Topic Highlight Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jul 7, 2016; 22(25): 5627-5641
Published online Jul 7, 2016. doi: 10.3748/wjg.v22.i25.5627
Liquid biopsy in patients with pancreatic cancer: Circulating tumor cells and cell-free nucleic acids
Taisuke Imamura, Shuhei Komatsu, Daisuke Ichikawa, Tsutomu Kawaguchi, Mahito Miyamae, Wataru Okajima, Takuma Ohashi, Tomohiro Arita, Hirotaka Konishi, Atsushi Shiozaki, Ryo Morimura, Hisashi Ikoma, Kazuma Okamoto, Eigo Otsuji, Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
Author contributions: Imamura T and Komatsu S equally contributed to this work; Imamura T and Komatsu S wrote the manuscript; Ichikawa D, Okamoto K and Otsuji E helped to draft the manuscript; Kawaguchi T, Miyamae M, Okajima W, Ohashi T, Arita T, Konishi H, Shiozaki A, Morimura R and Ikoma H collected the literatures.
Conflict-of-interest statement: The authors have no conflict of interest to report.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (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/
Correspondence to: Shuhei Komatsu, MD, PhD, Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto 602-8566, Japan. skomatsu@koto.kpu-m.ac.jp
Telephone: +81-75-2515527 Fax: +81-75-2515522
Received: March 27, 2016
Peer-review started: March 28, 2016
First decision: May 12, 2016
Revised: May 25, 2016
Accepted: June 15, 2016
Article in press: June 15, 2016
Published online: July 7, 2016

Abstract

Despite recent advances in surgical techniques and perioperative management, the prognosis of pancreatic cancer (PCa) remains extremely poor. To provide optimal treatment for each patient with Pca, superior biomarkers are urgently needed in all phases of management from early detection to staging, treatment monitoring, and prognosis. In the blood of patients with cancer, circulating tumor cells (CTCs) and cell-free nucleic acids (cfNAs), such as DNA, mRNA, and noncoding RNA have been recognized. In the recent years, their presence in the blood has encouraged researchers to investigate their potential use as novel blood biomarkers, and numerous studies have demonstrated their potential clinical utility as a biomarker for certain types of cancer. This concept, called “liquid biopsy” has been focused on as a less invasive, alternative approach to cancer tissue biopsy for obtaining genetic and epigenetic aberrations that contribute to oncogenesis and cancer progression. In this article, we review the available literature on CTCs and cfNAs in patients with cancer, particularly focusing on PCa, and discuss future perspectives in this field.

Key Words: Pancreatic cancer, Biomarker, Liquid biopsy, Circulating tumor cells, Cell-free nucleic acids

Core tip: In the blood of patients with cancer, circulating tumor cells (CTCs) and cell-free nucleic acids (cfNAs), such as DNA, mRNA, and noncoding RNA have been recognized. In the recent years, their presence in the blood has encouraged researchers to investigate their potential use as novel blood biomarkers. This concept, called “liquid biopsy” has been focused on as a less invasive, alternative approach to cancer tissue biopsy for obtaining genetic and epigenetic aberrations that contribute to oncogenesis and cancer progression. In this article, we review the available literature on CTCs and cfNAs in patients with cancer, particularly focusing on pancreatic cancer.



INTRODUCTION

Pancreatic cancer (PCa) is the fourth leading cause of cancer-related deaths in the United States, and the eighth worldwide[1]. In recent years, as a result of advances in surgical techniques and perioperative management, the perioperative mortality rate has decreased and perioperative chemotherapy and radiotherapy have greatly improved; however, prognostic outcomes for PCa remain poor[2,3]. Even now, the median survival time of patients with PCa is 5-8 mo and their 5-year survival rate is less than 10%[2,3].

Although surgical resection is the only option for macroscopic tumor clearance for PCa, most patients are diagnosed at an advanced and unresectable stage because PCa develops with no symptoms, local invasiveness, and metastases to distant organs in the early stage of its clinical course[1,4,5]. In addition, PCa shows resistance to conventional chemotherapies. Therefore, primary tumors must be detected at an early and resectable stage, whereas patients with far advanced disease must be preoperatively diagnosed to avoid surgical impairments and to select appropriate treatments to improve the quality of remaining life[6]. Consequently, to provide optimal management for each patient, biomarkers are urgently needed that are better than the conventional ones, such as carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9), in all phases of management from early detection to staging, treatment monitoring, and prognosis for PCa.

Numerous genetic and epigenetic aberrations contribute to oncogenesis and cancer progression, and the utility of these alterations for diagnostic, prognostic, and therapeutic purposes in various cancers have been investigated. Conventionally, these cancer-related alterations are investigated using tissue samples from surgical or biopsy specimens. These methodologies for pancreatic tissue acquisition cannot always be performed and repeated because of their invasive nature and anatomical difficulties. Thus, conventional examinations may fail to reflect current tumor dynamics and drug sensitivities, which may change during the therapeutic process. Detection and examination of circulating tumor cells (CTCs) and/or cell-free nucleic acids (cfNAs) in the bloodstream by performing a so-called liquid biopsy, which allows repeated sampling, makes it possible to track the current status of a tumor and its heterogeneous characteristics, which single sampling may fail to capture.

In the past decades, numerous studies have shown the potential utility of novel blood-based biomarkers, such as CTCs and cfNAs, for various cancers including PCa[7-10]. These promising markers are considered to possess great potential and could facilitate therapeutic strategies for cancer. In this article, we review the histological backgrounds, characteristics, and developments among CTCs and cfNAs in cancer research and discuss future perspectives, particularly focusing on PCa.

BIOLOGY AND DETECTION OF CTCS

In 1869, Ashworth[11] identified the presence of CTCs for the first time in the blood of a metastatic breast cancer patient in whom cells similar to those in the primary tumors were found in the blood at autopsy. Since then, many groups have challenged and demonstrated the identification and characterization of CTCs in peripheral blood of patients with cancer in several cancers. It has been recognized that CTCs originate from the primary tumor and/or metastatic lesions and are therefore extremely rare in healthy subjects and patients with nonmalignant diseases but are present in various metastatic carcinomas with a wide range of frequencies[12].

CTCs are generally thought to be quite heterogeneous in both phenotype and genotype, and only 2.5% of CTCs develop micrometastases and only 0.01% develop macrometastases[13-15]. During the journey toward the development of a metastatic lesion, some CTCs undergo epithelial-to-mesenchymal transition (EMT), which is characterized by decreased expression of epithelial markers and the acquisition of mesenchymal features[15] that could allow CTCs to escape from epithelial marker-based detection[16]. Furthermore, CTCs are present in peripheral blood at a low density among billions of blood cells in each milliliter of blood[17]. Consequently, accurate detection of CTCs with sufficient sensitivity and specificity has been a major technical challenge for researchers.

Currently, the CELLSEARCH system (Veridex) is the most widely used CTC platform. In this platform, immunomagnetic beads coated with anti-epithelial cellular adhesion molecule (EpCAM) antibodies capture CTCs, followed by immunostaining with two positive markers: cytokeratins (CKs) 8/18/19 for cytoplasmic epithelium and 4′,6′-diamidino-2-phenylindole hydrochloride for nucleic acids, and a negative marker, leukocyte-specific CD45. The CELLSEARCH system is the only CTC platform to gain the approval of the United States Food and Drug Administration, and its clinical utility has been demonstrated as a diagnostic and prognostic indicator in patients with metastatic breast, prostate, and colon cancer[18-23]. In contrast, EpCAM-based enrichment of CTCs such as the CELLSEARCH system could fail to capture CTCs that have undergone EMT and increase the malignant potential. Several problems still remain regarding the detection and isolation capability and, thus, the clinical utility of CTCs. To improve sensitivity and specificity despite the heterogeneity of CTCs, new technology for the isolation and enrichment of CTCs has been developed. More recently, CTC-Chip[24] was demonstrated to increase the detection of CTCs by using tumor-specific markers, such as PSA in prostate cancer or HER2 in breast cancer, in addition to epithelial markers. Furthermore, Saucedo-Zeni et al[25] reported a new technology that enables the capture and enrichment of CTCs in vivo using a medical Seldinger guidewire inserted through a standard venous cannula into the cubital veins. Despite these advances, however, the isolation and enrichment of CTCs remains at the development stage.

After the isolation and enrichment of CTCs, identification procedures must be performed to examine their genetic and biological features. Various methods, such as immunocytochemistry and molecular techniques, have been commonly performed for identifying CTCs. Conventionally, immunostaining using 4′,6′-diamidino-2-phenylindole hydrochloride as a nuclear stain, CK as an epithelial marker, and CD45 as a hematopoietic marker have been widely used[26]. Among molecular approaches, quantitative reverse transcription-polymerase chain reaction (RT-PCR) has been generally employed to investigate the molecular characteristics of CKs, CEA, and other driver markers[27].

CTC DETECTION IN PATIENTS WITH PCA AND ITS CLINICAL RELEVANCE

To date, many researchers have tried to detect CTCs in patients with PCa and have demonstrated its clinical utility using various approaches. Table 1 summarizes the previous reports about CTCs in patients with PCa. Early studies of CTCs in PCa employed tumor-specific and/or epithelial-related mRNAs as a molecular target for the detection of CTCs. Among these studies, RT-PCR techniques have been widely used for the detection of mRNAs despite a low concentration. Funaki et al[28] first reported the clinical utility of CTCs in patients with PCa using RT-PCR. They demonstrated that the detection of CEA mRNA as a tumor-specific molecule in peripheral blood is useful in finding the hematogenous spreading of adenocarcinoma cells. Three of nine patients with PCa (33.3%) were positive for CEA mRNA, and none of the control patients was positive for CEA mRNA in peripheral blood. Following this study, some groups evaluated the clinical utility of CEA mRNA in the bloodstream for the detection of CTCs in PCa and reported its sensitivity (47.8%-75.0%) and specificity (94.6%-96%) for the detection of PCa[29-32]. Chausovsky et al[33] reported that the epithelial-associated molecule CK-20 mRNA is useful for the detection of CTCs in PCa. They successfully demonstrated RT-PCR of CK-20 as a potential biomarker for detecting metastases in blood samples from patients with PCa and in subsequent studies reported data supporting this result[34-36]. Other mRNAs, such as epithelial growth factor receptor mRNA[37], α1,4-N-acetylglucosaminyltransferase mRNA[38], and CK-19 mRNA[39] have also been reported as useful targets for the detection of CTCs using RT-PCR in patients with PCa. Regarding these mRNA-based studies, sensitivities varied widely and were relatively low despite high specificity. Furthermore, some of these studies used mononuclear cell fraction from density gradient enrichment or blood samples without enrichment techniques to extract total RNA for the investigation of mRNA expression; therefore, the approach cannot exclude the possible contamination of leukocyte-originated RNAs, an issue that should be considered in interpreting results. More recently, Zhou et al[40] suggested that the combined detection of c-Met, h-TERT, CK20, and CEA using RT-PCR following immunomagnetic bead enrichment could be used as an indicator for circulating cancer cells with 100% sensitivity and 100% specificity in patients with PCa.

Table 1 Circulating tumor cells in pancreatic cancer.
Ref.Patient characteristicsNumber of patients with PCaControlsEnrichment/isolation methodDetection methodDetection rate
Funaki et al[28], 1996Pre- or post-treatment9NANoneRT-PCR: CEA mRNA33.3% of PCa
Funaki et al[29], 1998Preoperative3NANoneRT-PCR: CEA-mRNA
Miyazono et al[30], 1999Intra-operative2115 HVDensity gradient enrichmentRT-PCR: CEA-mRNA61.9% of PCa
Chausovsky et al[33], 1999Metastatic PCa2822 BDDensity gradient enrichmentRT-PCR: CK20 mRNA78.6% of PCa
Z'graggen et al[124], 2001All stages, preoperative10566 HVDensity gradient enrichmentICC: CK-AE1/AE3Sensitivity: 26%, specificity: 96%
Lukyanchuk et al[34], 2003NA1118 HVDensity gradient enrichmentRT-PCR: CK-20 and PSCACK-20: 19 of 47 (40.4%), PSCA: 22 of 47 (46.8%)
Clarke et al[37], 2003NA1123 HVDensity gradient enrichmentRT-PCR: EGFR mRNA18% of PCa, 0.0% of HV
Mataki et al[32], 2004All stages2015 HV, 15 BDDensity gradient enrichmentRT-PCR: CEA mRNASensitivity: 75.0%, specificity: 94.6%
Zhang et al[35], 2005Stages II and III405 BD, 5 HVNot availableRT-PCR: CK20 mRNA57.5% of PCa
Soeth et al[36], 2005Preoperative154NADensity gradient enrichmentRT-PCR: CK20 mRNA33.8% of PCa
Ishizone et al[38], 2006All stages5510 CP, 70 HVDensity gradient enrichmentRT-PCR: a4GnT76.4% of PCa, 40.0% of CP, 17.1% of HV
Hoffmann et al[39], 2007All stages3716 CP, 15 BDDensity gradient enrichmentRT-PCR: CK-19 mRNA64% of PCa
Kurihara et al[41], 2008Stages II, III and IV2611 CP, 10 HVCELLSEARCH®42% of PCa
Zhou et al[40], 2011All stages2515 BDImmunomagnetic separationRT-PCR: C-MET, h-TERT, CK20, and CEASensitivity: 100%, specificity: 93.3%
Khoja et al[125], 2012Stages III and IV54 PCaNASize-based selectionRT-PCR: EpCAM, CK, vimentin, and CEAISET 49/54 (93%) vs CELLSEARCH® 21/54 (40%)
de Albuquerque et al[126], 2012Stages III and IV3440 HVImmunomagnetic separationRT-PCR: KRT19, MUC1, EpCAM, CEACAM5, and BIRC5Sensitivity: 47.1%, specificity: 100%
Kamande et al[127], 2013All stages125 HVMicrofluidic; ICCDAPI+, CD45-, CK+100% of PCa
Bidard et al[42], 2013Locally advanced PCa79NACELLSEARCH®11% of PCa
Iwanicki-Caron et al[43], 2013All stages40NASize-based selectionCell size and cytopathologic criteriaSensitivity: 55.5%, specificity: 100%, accuracy: 70%
Bobek et al[45], 2014All stages24NASize-based selectionDAPI, CK, CEA, vimentin IHC66.7% of PCa
Sheng et al[48], 2014Metastatic PCa18NAMultifluidic, "GEM"Chip94.4% of PCa
Rhim et al[47], 2014All stages1119 HVGeometrically enhanced differential immunocaptureICC for DAPI, CD45, CK, and PDX-173% of PCa
Catenacci et al[128], 2015Stages II, III and IV18NAImmune-magnetic separationCD45-negative and positive for CK8, -18, and/or -19 and DAPI118.4 ± 36.8 CTCs/7.5 mL PVB, compared with a mean of 0.8 ± 0.4 CTCs/7.5 mL PB
Earl et al[81], 2015All stages35NACELLSEARCH®20% of PCa
Cauley et al[44], 2015All stages1059 HVSize-based selectionCytomorphologic criteria48.6% of PCa
Kulemann et al[46], 2015Preoperative119 HVSize-based selection: ScreenCellCytologic and detection of KRAS mutation75% of early PCa, 71.4% of advanced PCa
Zhang et al[129], 2015All stages3230 HVICC and FISHDAPI+, CD45-, and CK+, or CEP8 > 2+Sensitivity: 63.6%, specificity: 94.4%
Bissolati et al[130], 2015Intra-operative20NACELLSEARCH®PVB: 40%, PB: 20%
Zhang et al[131], 20151515 HVImmunomagnetic separationBC-15 aptamer or anti-CK staining73.3% of PCa

Meanwhile, an immunocytochemical approach also has been used for identifying CTCs in patients with PCa. Although many studies have investigated the clinical utility of the CELLSEARCH platforms in PCa, a low detection rate ranging from 5% to 42% was reported[12,41,42]. In contrast, some of the studies demonstrated the clinical utility of CTCs as a predictor of disease recurrence and prognosis by reporting that a positive CTC finding was associated with disease recurrence and/or poor survival. To improve the detection rate of the CELLSEARCH system in PCa, several researchers have developed novel technologies for the detection of CTCs, such as size-based isolation[43-46] and microfluidics[47,48], and reported favorable results.

Overall, a certain level of utility of CTCs as a biomarker in PCa may be practically guaranteed; however, there are several problems that remain to be solved before CTCs can be considered useful in a clinical setting. First, CTCs might not be suitable in screening tests for early detection (which is desirable in PCa) owing to their low sensitivity. Novel technology with high sensitivity for the detection of CTCs is required to use CTCs in screening tests for early diagnosis. Second, the detection of CTCs has to overcome the challenge of the rarity and heterogeneity of CTCs, and because the methodology for the detection of CTCs remains in a developmental stage, the approach to CTCs and the results of studies have varied widely. Consequently, it is difficult to assess, compare, and interpret the results of multiple studies and establish the evidence and clinical relevance of CTCs. Establishment of a unified methodology and large-scale validation of their utility are required for wider clinical application.

BIOLOGY AND DETECTION OF CFNAS

For many decades, cfNAs have been known to be present in peripheral blood. Mandel and Metais[49] first reported that nucleic acids were detectable in human plasma in 1948. In 1977, Leon et al[50] detected cell-free DNAs in the serum of patients with cancer. Since then, several studies have identified tumor-specific and/or tumor-associated alterations in the circulating cfNAs of patients with various cancers. In 1989, Vasioukhin et al[51] successfully detected cell-free DNA with neoplastic characteristics, providing the first evidence suggesting that tumors can shed DNA into the circulation. This hypothesis was further supported by a study in which a NRAS mutation in the plasma of patients with myelodysplastic syndrome or acute myelogenous leukemia[52] and KRAS mutation in the plasma or serum of patients with PCa were detected[53].

Cell-free RNA, which is thought to be more fragile than DNA because RNA is easily degraded by endogenous ribonuclease (RNase) in plasma/serum, has been successfully detected in blood. In 1999, tyrosinase mRNA in the serum of patients with malignant melanoma[54] and Epstein-Barr virus-associated RNA in nasopharyngeal carcinoma[55] was successfully detected. Subsequently, many studies have demonstrated the presence of specific mRNA in plasma/serum and its clinical utility in patients with various cancers[56-58].

Regarding noncoding RNA, Mitchell et al[59] firstly demonstrated that circulating microRNAs (miRNAs) had the potential to be novel biomarkers in patients with solid cancers in 2008. Since then, numerous studies examining circulating noncoding RNAs have been performed, with most studies focusing on miRNAs. Although other noncoding RNAs, such as small nucleolar RNA (snoRNA), small nuclear RNA (snRNA), piwi-interacting RNA (piRNA), and long noncoding RNA (lncRNA), have been recognized to have biological functions and may have great potential to be novel blood biomarkers, there are few reports of these noncoding RNAs. Further studies and accumulation of evidence are required.

CIRCULATING CELL-FREE DNA IN PLASMA/SERUM AND PCA

The study of circulating cell-free DNA in the plasma/serum involves two major strategies: the measurement of the amount of cell-free DNA in the circulation and the detection of tumor-derived genetic aberrations such as point mutations, allelic imbalances, microsatellite instability, genetic polymorphisms, loss of heterozygosity, and methylation. Of these, many reports have demonstrated the detection of genetic and epigenetic alterations in circulating cell-free DNA in the bloodstream of patients with cancer[60-63].

Previous reports about circulating cell-free DNA in PCa are summarized in Table 2. In 1983, Shapiro et al[64] first reported the presence of circulating cell-free DNA in PCa. This study demonstrated that serum DNA concentration is markedly elevated in patients with PCa compared with normal controls using radioimmunoassay and that an abnormally high concentration of DNA in serum may have diagnostic and prognostic value. Since then, investigations of tumor-derived genetic alterations in plasma/serum have become mainstream. Only a few studies investigating the methylation[65,66], microsatellite instability, and allelic imbalance[67] can be found. In contrast, the detection of K-ras mutation in plasma/serum appears to be the most widely used approach in the diagnosis of PCa. It has been widely recognized that over 90% of pancreatic adenocarcinomas contain mutated K-ras genes[68-70], and the detection of mutant K-ras provides a definitive diagnosis of pancreatic adenocarcinoma in pancreatic tissue[70,71]. Based on the detection of K-ras mutation in tumor tissues, the detection of the tumor-derived genetic alterations in circulating cell-free DNA has been attempted for the diagnosis of PCa. In 1993, Yamada et al[72] demonstrated that detection of K-ras mutations in plasma may be clinically useful for evaluating tumor burden and efficacy of treatment for PCa. Subsequently, many groups have reported the possible clinical utility of K-ras mutations in circulating cell-free DNA in PCa[73-80].

Table 2 Circulating cell-free DNA in pancreatic cancer.
Ref.PatientsControlsSampleMethodTarget candidateFindings
Shapiro et al[64], 1983201 MD185 BDSerumRadioimmunoassayCirculating DNA levelsSerum DNA concentration is markedly elevated in 90% of patients with PCa as compared with HV
Yamada et al[72], 199821 PCa-PlasmaMutant allele-specific amplification methodK-ras mutationIn 9 of 15 (60%) patients with K-ras gene mutation-positive tumors, an identical mutation was detected in the plasma DNA. Detection of K-ras mutations in plasma may be clinically useful for evaluating tumor burden and efficacy of treatment
Giacona et al[132], 19983 PCa3 HVPlasmaGel electrophoresis and measuring the variation in length by electron microscopyLengthThere are significant differences in non-cell-associated DNA in plasma between patients with PCa and HV
Theodor et al[73], 199920 PCa6 CP, 5 HVSerumPCRK-ras mutationK-ras gene mutations at codon 12 were detected in the sera of 14 of 20 patients with PCa and in none of the 6 patients with CP, or in the 5 HVs
Castells et al[74], 199944 PCa37 CPPlasmaRestriction fragment length polymorphism-PCR and single-strand conformation polymorphism techniquesK-ras mutationPlasma K-ras analysis is a highly specific, low-sensitivity approach that has diagnostic and prognostic clinical implications in patients with PCa
Zambon et al[75], 200029 PCa12 HVSerumME-PCRK-ras mutationK-ras was amplifiable in 2 patients with PCa (6.9%), and K-ras was not amplifiable in any of the 12 serum samples obtained from HVs
Maire et al[76], 200247 PCa31 CPSerumPCR and allele-specific amplificationKRAS2 mutationsKRAS2 mutations were found in 22 patients (47%) with PCa and in 4 controls with CP (13%) (P < 0.002)
Melnikov et al[65], 200930 PCa30 HVPlasmaMultiplexed array-mediated analysis of DNA methylationMethylationDifferential methylation profiling of plasma DNA can detect PCa with 76% sensitivity and 59% specificity
Liggett et al[66], 201030 PCa30 CP, 30 HVPlasmaMicroarray-mediated methylation analysisMethylationMethylation analysis achieved 81.7% sensitivity and 78% specificity (P < 0.01) in the detection of CP (HV vs CP) and 91.2% sensitivity and 90.8% specificity (P < 0.01) in the differential detection of PCa (PCa vs CP)
Chen et al[79], 201091 PCa-PlasmaDirect sequencingK-rasK-ras codon 12 mutations were found in 30 of 91(33%) plasma DNA samples and significantly reflected the clinical parameters, including TNM tumor staging and liver metastasis, and independent predict shorter survival time
Wu et al[80], 201424 PCa25 HVPlasmaCOLD-PCR combined with an unlabeled-probe HRM approachK-rasKRAS mutations were identified in 26 of 36 PCa cases (72.2%), but none were detected in the disease control and/or healthy group
Earl et al[81], 201531 PCa-PlasmaDigital PCRKRASKRAS mutant cfDNA was detected in 26% of patients at all stages, which correlated strongly with OS, 60 d for KRAS mutation-positive vs 772 days for KRAS mutation-negative patients
Zill et al[85], 201526 PCa-PlasmaSequenced on an Illumina Hi-Seq 2500KRAS, TP53, APC, FBXW7, and SMAD4The diagnostic accuracy of cfDNA sequencing was 97.7%, with 92.3% average sensitivity and 100% specificity across 5 informative genes
Singh et al[133], 2015PlasmaLevels of ctDNA and K-ras mutationHigher levels of plasma DNA were significantly associated with lower OS and advanced stage. However, k-ras mutation did not correlate with any of the clinicopathological parameters or survival
Kinugasa et al[82], 2015141 PCa20 CP, 20 HVSerumDigital PCRG12V, G12D, and G12R in codon 12 of K-ras geneK-ras mutation rate in ctDNA was 62.6%. The survival of patients with K-ras mutations in ctDNA was significantly shorter than that of patients without mutations
Sausen et al[83], 201577 PCa-PlasmaNext-generation sequencingThe 43% of patients with localized disease had detectable ctDNA at diagnosis. Detection of ctDNA after resection predicts clinical relapse and poor outcome, with recurrence by ctDNA detected 6.5 months earlier than with CT imaging
Takai et al[84], 2015259 PCa-PlasmaPicoliter-droplet digital PCR and targeted deep sequencingKRAS mutationKRAS mutations were identified in 14 of 48 patients (29.2%) examined by targeted deep sequencing of cfDNA

PCR has been widely used to detect tumor-derived mutations in genes isolated from the serum and plasma of patients with cancer[51,53]. More recently, droplet digital PCR[81-84] and genome-wide high-throughput sequencing[84,85] has been demonstrated as a potential detection tool for rare mutations and multiple types of mutations in circulating DNA with great accuracy. Using these new technologies in several solid cancers such as colorectal, breast, and ovarian cancer, a correlation has been found between the acquisition of drug resistance and genetic aberrations in cell-free DNA in the blood of patients under treatment[86-88]. It is hoped that the potent utility of cell-free DNA for the assessment of residual disease, recurrence, and acquisition of drug resistance as well as for the detection of disease can be proven in PCa.

CIRCULATING CELL-FREE MRNA IN PLASMA/SERUM AND PCA

Although RNA is easily degraded by RNase and the concentration of RNase in blood is high in patients with cancer[89], many groups have demonstrated the stable presence of cell-free mRNAs in the blood of patients with cancer. Recently, it has been considered that these RNAs could be incorporated into exosomes, microvesicles, and multivesicles, which seem to be adequately protected against the degradation caused by the abundant RNases and released from the cellular surface to the bloodstream[90]. There are numerous studies investigating the correlation between cell-free mRNA in the bloodstream and several solid cancers, and these studies are mainly aimed at investigating the mRNAs in plasma/serum that are up-regulated in cancer tissues[56-58,91-93]. Regarding PCa, there are several studies investigating mRNA in peripheral blood mononuclear cells and CTCs[29,32,35,38] as a marker for the detection of CTCs; however, the number of cell-free mRNAs in plasma/serum is extremely small. Kang et al[94] recently demonstrated that type IV collagen (COL6A3) mRNA in serum might serve as a biomarker for the detection of PCa according to tumor-specific alternative splicing. Accumulation of evidence and understanding of cell-free mRNA in patients with cancer may have a potential to bring new insights into the field of liquid biopsy in PCa.

CIRCULATING NONCODING RNA IN PLASMA/SERUM

It has been revealed that as much as 80% of genomic DNA is transcribed into RNAs[95]. In contrast, the Human Genome Project discovered that the open reading frames of protein genes constitute less than 2% of the 3.2 billion bases[96,97]. Thus, a large portion of human genomic DNA does not code proteins. It is now becoming evident that a variety of noncoding RNAs (ncRNAs) play important roles in many cellular processes and are not just mere intermediates in the transfer of genetic information from DNA to proteins, which indicates that the ncRNAs expression patterns could be used as molecular markers in specific diagnostic methods[98].

For circulating ncRNAs, almost all of the studies have focused on miRNAs, which are short noncoding RNAs that play important roles in various physiologic and developmental processes. One strand (a guide strand) of mature miRNA is then incorporated into the RNA-induced silencing complex and subsequently hybridizes to the 3′-untranslated region of their target mRNAs to repress translation or degrade these mRNAs. Thus, a single miRNA can influence the expression of hundreds of genes and allow them to function in a coordinated manner. Therefore, miRNAs have been implicated as key molecules in all cellular processes. Numerous studies have shown that alterations in miRNA expression correlate with various diseases, including the development and progression of cancer, and some miRNAs can function as oncogenes or tumor suppressors. Furthermore, several recent studies have demonstrated that some extracellular miRNAs occur not only through cell lysis but also through active secretion[8,99,100]. Cell-derived endogenous miRNAs are present in the blood in a remarkably stable form that is protected from endogenous RNase activity. Kosaka et al[100] demonstrated that a subset of miRNAs can be packaged into exosome vesicles and released through a ceramide-dependent secretory mechanism. Furthermore, most miRNAs are stable in plasma owing to their binding to proteins such as Argonaute2 and high-density lipoprotein[101]. All circulating miRNAs, regardless of whether they are incorporated into protein complexes and/or cell-derived microvesicles, seem to be adequately protected against the degradation caused by the abundant RNases in human plasma and serum. These findings have opened up a new and interesting field in the diagnosis of cancer and the treatments of patients with cancer.

Mitchell et al[59] first demonstrated that circulating miRNAs have the potential to be novel blood biomarkers in patients with solid cancers in 2008. Since then, numerous studies of circulating miRNAs in cancer have been performed to investigate their potential as candidate novel biomarkers. Regarding PCa, previous reports about circulating miRNAs are summarized in Table 3. Wang et al[102] first reported the plasma level of four candidate miRNAs (miR-21, -155, -196a, and -210) that were previously reported to be up-regulated in PCa tissue. Since then, several groups have reported the utility of circulating miRNAs as biomarkers for PCa, and the number of studies and the variety of miRNAs have been increasing. Many studies have demonstrated the clinical significance of deregulated expression of miRNA in plasma/serum, and more than 30 miRNAs have been reported as candidate novel blood biomarkers in PCa. As mentioned before, one miRNA can regulate multiple mRNAs and the numbers of discovered miRNAs and targeted mRNAs are still increasing owing to recent advances in analysis technology. Consequently, more recently, diagnosing PCa with higher sensitivity and specificity has been attempted by employing multiple miRNAs[103,104]. Schultz et al[103] reported that two diagnostic panels including four (miR-145, miR-150, miR-223, and miR-636) and 10 (miR-26b, miR-34a, miR-122, miR-126*, miR-145, miR-150, miR-223, miR-505, miR-636, and miR-885.5p) miRNAs based on the expression in whole blood could be used to detect PCa with high sensitivity and specificity. However, it should be considered that the study employed whole blood as a sample for extracting RNAs; therefore, the miRNAs found obviously included miRNAs derived from blood cells or CTCs other than cell-free miRNAs. Kojima et al[104] reported that a combination of eight miRNAs (miR-6075, miR-4294, miR-6880-5p, miR-6799-5p, miR-125a-3p, miR-4530, miR-6836-3p, and miR-4476) could achieve high sensitivity, specificity, and accuracy for the detection of PCa.

Table 3 Circulating noncoding RNA in pancreatic cancer.
Ref.SampleCandidate targetPotential value
miRNA
Wang et al[102], 2009PlasmamiR-210 (↑), miR-21 (↑), miR-155 (↑), miR-196a (↑)D
Ho et al[134], 2010PlasmamiR-210 (↑)D
Li et al[135], 2010PlasmamiR-200a (↑), miR-200b (↑)D
Ali et al[136], 2010PlasmamiR-21 (↑)D/P
Kong et al[105], 2011SerummiR-196a (↑)D/S/P
LaConti et al[137], 2011SerummiR-155 (↑)D
Morimura et al[138], 2011PlasmamiR-18a (↑)D
Liu et al[139], 2012SerummiR-16 (↑), miR-196a (↑)D
Liu et al[140], 2012SerummiR-20a (↑), miR-21 (↑), miR-24 (↑), miR-25 (↑),D/P
miR-99a (↑), miR-185 (↑), miR-191 (↑)
Li et al[141], 2012SerummiR-1290 (↑)D
Wang et al[142], 2013Whole bloodmiR-27a-3p (↑)D
Kawaguchi et al[143], 2013PlasmamiR-221 (↑), miR-375 (↓)D/S
Zhao et al[144], 2013SerummiR-192 (↑)D
Li et al[141], 2013SerummiR-1290 (↑)D
Wang et al[145], 2013SerummiR-21 (↑)T/P
Carlsen et al[146], 2013PlasmamiR-375 (↑)D
Que et al[147], 2013SerummiR-17-5p (↑), miR-21 (↑), miR-155 (↓), miR-196a (↓)D/S
Schultz et al[103], 2014Whole bloodMultigene indexD
Gao et al[148], 2014PlasmamiR-16 (↑)D
Chen et al[149], 2014PlasmamiR-182 (↑)D/S/P
Ganepola et al[150], 2015PlasmamiR-22 (↑), miR-642b (↑), miR-885-5p (↑)D
Abue et al[151], 2015PlasmamiR-21 (↑), miR-483-3p (↑)D/S/P
Slater et al[152], 2015SerummiR-196a (↑), miR-196b (↑)D
Kojima et al[104], 2015SerumMultigene indexD
Xu et al[153], 2015PlasmamiR-486-5p (↑), miR-938 (↑)D
Madhavan et al[154], 2015SerummiR-1246 (↑), miR-3976 (↑), miR-4306 (↑), miR-4644 (↑)D
Komatsu et al[123], 2015PlasmamiR-223 (↑)D/P
Miyamae et al[106], 2015PlasmamiR-744 (↑)D/S/P/T
Other noncoding RNAs
Wang et al[117], 2015PlasmaHOTTIP-005 (↑), RP11-567G11.1 (↑)D/P
Baraniskin et al[155], 2013Plasma/serumU2 snRNA (↑)D

Furthermore, the studies of circulating cell-free miRNAs in PCa have demonstrated the usefulness of circulating miRNAs as a staging marker, treatment marker, and prognostic marker as well as a biomarker for the detection of PCa. Kong et al[105] demonstrated that the serum miR-196a expression level had potential value for predicting median survival time of patients with PCa [high-level miR-196a, 6.1 mo (95%CI: 4.49-7.72), vs low-level miR-196a, 12.00 mo, (95%CI: 5.92-18.08), P = 0.007). Most recently, our group demonstrated that high expression of miR-744 in plasma might be a useful biomarker for screening PCa, monitoring, and predicting poor prognosis and chemoresistance in patients with PCa[106]. Although more evidence has been accumulating as we have been reviewing previous reports, several problems remain be solved for clinical application. There is no consensus regarding inter- and intra-individual variation, whether plasma or serum is more appropriate, and what molecule is optimal for the most sensitive detection and endogenous controls.

Some kinds of noncoding RNAs other than miRNA have been recognized to have biological functions. Especially in cancers, some noncoding RNAs have been demonstrated to have oncogenic or tumor-suppressive functions and to be deregulated in tumor tissue. Regarding PCa, HOTAIR[107], MALAT-1[108], MEG3[109], Gas5[110], HULC[111], PVT1[112], PPP3CB, MAP3K1, DAPK1[113], BC008363[114], ENST00000480739[115], and HSATII[116] have been reported to have tumor-associated functions and tumor-specific expression. However, there are no studies investigating these cell-free RNAs in the bloodstream of patients with PCa. The noncoding RNAs other than miRNAs, such as long noncoding RNA (lncRNA), small nucleolar RNA (snoRNA), small nuclear RNA (snRNA), and Piwi-interacting RNA (piRNA) in the bloodstream of patients with PCa remain largely unexplored. Most recently, Wang et al[117] demonstrated that the plasma fragments of lncRNA, HOTTIP-005, and RP11-567G11.1 have the potential to be used as diagnostic biomarkers of PCa. We believe that future studies of circulating noncoding RNAs in PCa will bring new insights to this field.

CURRENT ISSUES AND FUTURE PERSPECTIVE

Blood-based biomarkers, evaluated using liquid biopsy, are attractive as diagnostic, staging, prognostic, and treatment markers for PCa owing to their less invasive nature, and the clinical relevance of using liquid biopsy to identify biomarkers in PCa has become practically guaranteed. However, several problems remain to be solved before application in a clinical setting. One of the important hurdles to overcome is the lack of consensus regarding a technical approach. The methodology for the detection and assessment of CTCs and circulating cfNAs remains in a developmental stage, and therefore, the techniques used, such as sample type, storage conditions, target molecules, and detection approach, have varied widely among research groups. The standardization and unification of techniques through all processes and the accumulation of many results under the same conditions or in large-scale studies should be emphasized. Meanwhile, recent technical advances allow us to detect a slight amount of circulating cell and nucleic acids even in the body fluid other than blood, and several recent studies have already reported the possible utility of CTC and cfNAs in body fluids other than blood[118-122]. These reports have suggested the possibility of even less invasive and even more effective biomarkers in near future.

For the development of the field, PCa seems to be the ideal cancer to investigate for several reasons. First, the investigation of tumor-associated genetic alterations using tissue samples obtained from surgical or biopsy specimens is costly and difficult owing to anatomical and clinical difficulties. Additionally, the utility of the current serum biomarkers is limited owing to insufficient sensitivity and specificity. Repeatable and less invasive testing with high sensitivity and specificity to obtain information about genetic alterations or tumor dynamics will considerably contribute to the improvement of management for PCa. Second, it has already been revealed that some genetic or epigenetic alterations, such as KRAS, p16, SMAD4, TP53, and CDKN2A are present in most pancreatic tumor cells, which might make it easy to confirm the targets as tumor-derived molecules. Third, a genetic evolutionary model[120] revealed that 10-30 years are required from initiating a mutation until a patient’s death. Furthermore, mucinous cystic neoplasms, intraductal papillary neoplasms, pancreatic intraepithelial neoplasia (IPMN), and intraductal tubular papillary neoplasms were identified as premalignant lesions of PCa[121] that develop to invasive PCa through stepwise progression with the accumulation of several genetic aberrations. If these important genetic aberrations could be captured by liquid biopsy, screening and monitoring tests for high-risk lesions or early detection could be realized. To date, there are few reports about the usefulness of liquid biopsy in these premalignant lesions of PCa[122]. Recently our study successfully demonstrated that plasma miR-223 could predict malignant potential of IPMN[123]. We believe that further studies of liquid biopsy in premalignant lesions of PCa could contribute to improve the prognostic outcomes of PCa patients and the biomarker for premalignant lesion is nearing the clinical application.

Overall, liquid biopsy has the potential to allow us to diagnose at an early stage, predict prognosis, track the current status such as therapeutic efficacy or resistance, and provide optimal, individual treatment strategies for patients with cancer, that is, tailor-made treatment. The development of liquid biopsy could provide many benefits for patients with cancer, especially for patients with PCa.

Footnotes

Manuscript Source: Invited manuscript

Specialty Type: Gastroenterology and Hepatology

Country of Origin: Japan

Peer-Review Report Classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C, C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Sawas T, Sica GS, Sperti C S- Editor: Ma YJ L- Editor: A E- Editor: Wang CH

References
1.  Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 2009. CA Cancer J Clin. 2009;59:225-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7953]  [Cited by in F6Publishing: 8022]  [Article Influence: 534.8]  [Reference Citation Analysis (2)]
2.  Stathis A, Moore MJ. Advanced pancreatic carcinoma: current treatment and future challenges. Nat Rev Clin Oncol. 2010;7:163-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 555]  [Cited by in F6Publishing: 603]  [Article Influence: 43.1]  [Reference Citation Analysis (0)]
3.  Wolfgang CL, Herman JM, Laheru DA, Klein AP, Erdek MA, Fishman EK, Hruban RH. Recent progress in pancreatic cancer. CA Cancer J Clin. 2013;63:318-348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 572]  [Cited by in F6Publishing: 636]  [Article Influence: 57.8]  [Reference Citation Analysis (0)]
4.  Heinemann V, Boeck S, Hinke A, Labianca R, Louvet C. Meta-analysis of randomized trials: evaluation of benefit from gemcitabine-based combination chemotherapy applied in advanced pancreatic cancer. BMC Cancer. 2008;8:82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 296]  [Cited by in F6Publishing: 334]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
5.  Sultana A, Tudur Smith C, Cunningham D, Starling N, Neoptolemos JP, Ghaneh P. Meta-analyses of chemotherapy for locally advanced and metastatic pancreatic cancer: results of secondary end points analyses. Br J Cancer. 2008;99:6-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 79]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
6.  Ozaka M, Matsumura Y, Ishii H, Omuro Y, Itoi T, Mouri H, Hanada K, Kimura Y, Maetani I, Okabe Y. Randomized phase II study of gemcitabine and S-1 combination versus gemcitabine alone in the treatment of unresectable advanced pancreatic cancer (Japan Clinical Cancer Research Organization PC-01 study). Cancer Chemother Pharmacol. 2012;69:1197-1204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 66]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
7.  Alix-Panabières C, Pantel K. Circulating tumor cells: liquid biopsy of cancer. Clin Chem. 2013;59:110-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 748]  [Cited by in F6Publishing: 795]  [Article Influence: 72.3]  [Reference Citation Analysis (2)]
8.  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)]
9.  van de Stolpe A, Pantel K, Sleijfer S, Terstappen LW, den Toonder JM. Circulating tumor cell isolation and diagnostics: toward routine clinical use. Cancer Res. 2011;71:5955-5960.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 154]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
10.  Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol. 2013;10:472-484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1114]  [Cited by in F6Publishing: 1199]  [Article Influence: 109.0]  [Reference Citation Analysis (0)]
11.  Ashworth TR. A case of cancer in which cells similar to those in the tumours were seen in the blood after death. Aust Med J. 1869;14:146-149.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Allard WJ, Matera J, Miller MC, Repollet M, Connelly MC, Rao C, Tibbe AG, Uhr JW, Terstappen LW. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res. 2004;10:6897-6904.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1810]  [Cited by in F6Publishing: 1867]  [Article Influence: 98.3]  [Reference Citation Analysis (0)]
13.  Fidler IJ. Metastasis: quantitative analysis of distribution and fate of tumor emboli labeled with 125 I-5-iodo-2’-deoxyuridine. J Natl Cancer Inst. 1970;45:773-782.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  O'Flaherty JD, Gray S, Richard D, Fennell D, O’Leary JJ, Blackhall FH, O’Byrne KJ. Circulating tumour cells, their role in metastasis and their clinical utility in lung cancer. Lung Cancer. 2012;76:19-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 121]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
15.  Luzzi KJ, MacDonald IC, Schmidt EE, Kerkvliet N, Morris VL, Chambers AF, Groom AC. Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am J Pathol. 1998;153:865-873.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 826]  [Cited by in F6Publishing: 820]  [Article Influence: 31.5]  [Reference Citation Analysis (0)]
16.  Gorges TM, Tinhofer I, Drosch M, Röse L, Zollner TM, Krahn T, von Ahsen O. Circulating tumour cells escape from EpCAM-based detection due to epithelial-to-mesenchymal transition. BMC Cancer. 2012;12:178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 393]  [Cited by in F6Publishing: 405]  [Article Influence: 33.8]  [Reference Citation Analysis (0)]
17.  Balic M, Lin H, Williams A, Datar RH, Cote RJ. Progress in circulating tumor cell capture and analysis: implications for cancer management. Expert Rev Mol Diagn. 2012;12:303-312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 44]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
18.  Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, Reuben JM, Doyle GV, Allard WJ, Terstappen LW. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;351:781-791.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3360]  [Cited by in F6Publishing: 3257]  [Article Influence: 162.9]  [Reference Citation Analysis (0)]
19.  Danila DC, Heller G, Gignac GA, Gonzalez-Espinoza R, Anand A, Tanaka E, Lilja H, Schwartz L, Larson S, Fleisher M. Circulating tumor cell number and prognosis in progressive castration-resistant prostate cancer. Clin Cancer Res. 2007;13:7053-7058.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 517]  [Cited by in F6Publishing: 497]  [Article Influence: 31.1]  [Reference Citation Analysis (0)]
20.  Hayes DF, Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Miller MC, Matera J, Allard WJ, Doyle GV, Terstappen LW. Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival. Clin Cancer Res. 2006;12:4218-4224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 765]  [Cited by in F6Publishing: 752]  [Article Influence: 44.2]  [Reference Citation Analysis (0)]
21.  Riethdorf S, Fritsche H, Müller V, Rau T, Schindlbeck C, Rack B, Janni W, Coith C, Beck K, Jänicke F. Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: a validation study of the CellSearch system. Clin Cancer Res. 2007;13:920-928.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 996]  [Cited by in F6Publishing: 973]  [Article Influence: 57.2]  [Reference Citation Analysis (0)]
22.  Sastre J, Maestro ML, Puente J, Veganzones S, Alfonso R, Rafael S, García-Saenz JA, Vidaurreta M, Martín M, Arroyo M. Circulating tumor cells in colorectal cancer: correlation with clinical and pathological variables. Ann Oncol. 2008;19:935-938.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 170]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
23.  Shaffer DR, Leversha MA, Danila DC, Lin O, Gonzalez-Espinoza R, Gu B, Anand A, Smith K, Maslak P, Doyle GV. Circulating tumor cell analysis in patients with progressive castration-resistant prostate cancer. Clin Cancer Res. 2007;13:2023-2029.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in F6Publishing: 269]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
24.  Stott SL, Hsu CH, Tsukrov DI, Yu M, Miyamoto DT, Waltman BA, Rothenberg SM, Shah AM, Smas ME, Korir GK. Isolation of circulating tumor cells using a microvortex-generating herringbone-chip. Proc Natl Acad Sci USA. 2010;107:18392-18397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1247]  [Cited by in F6Publishing: 1167]  [Article Influence: 83.4]  [Reference Citation Analysis (0)]
25.  Saucedo-Zeni N, Mewes S, Niestroj R, Gasiorowski L, Murawa D, Nowaczyk P, Tomasi T, Weber E, Dworacki G, Morgenthaler NG. A novel method for the in vivo isolation of circulating tumor cells from peripheral blood of cancer patients using a functionalized and structured medical wire. Int J Oncol. 2012;41:1241-1250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 135]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
26.  Lustberg MB, Balasubramanian P, Miller B, Garcia-Villa A, Deighan C, Wu Y, Carothers S, Berger M, Ramaswamy B, Macrae ER. Heterogeneous atypical cell populations are present in blood of metastatic breast cancer patients. Breast Cancer Res. 2014;16:R23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 76]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
27.  Pantel K, Alix-Panabières C, Riethdorf S. Cancer micrometastases. Nat Rev Clin Oncol. 2009;6:339-351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 488]  [Cited by in F6Publishing: 528]  [Article Influence: 35.2]  [Reference Citation Analysis (0)]
28.  Funaki NO, Tanaka J, Kasamatsu T, Ohshio G, Hosotani R, Okino T, Imamura M. Identification of carcinoembryonic antigen mRNA in circulating peripheral blood of pancreatic carcinoma and gastric carcinoma patients. Life Sci. 1996;59:2187-2199.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Funaki NO, Tanaka J, Hosotani R, Kogire M, Suwa H, Imamura M. Quantitative analysis of carcinoembryonic antigen messenger RNA in peripheral venous blood and portal blood of patients with pancreatic ductal adenocarcinoma. Clin Cancer Res. 1998;4:855-860.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Miyazono F, Takao S, Natsugoe S, Uchikura K, Kijima F, Aridome K, Shinchi H, Aikou T. Molecular detection of circulating cancer cells during surgery in patients with biliary-pancreatic cancer. Am J Surg. 1999;177:475-479.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Uchikura K, Takao S, Nakajo A, Miyazono F, Nakashima S, Tokuda K, Matsumoto M, Shinchi H, Natsugoe S, Aikou T. Intraoperative molecular detection of circulating tumor cells by reverse transcription-polymerase chain reaction in patients with biliary-pancreatic cancer is associated with hematogenous metastasis. Ann Surg Oncol. 2002;9:364-370.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Mataki Y, Takao S, Maemura K, Mori S, Shinchi H, Natsugoe S, Aikou T. Carcinoembryonic antigen messenger RNA expression using nested reverse transcription-PCR in the peripheral blood during follow-up period of patients who underwent curative surgery for biliary-pancreatic cancer: longitudinal analyses. Clin Cancer Res. 2004;10:3807-3814.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
33.  Chausovsky G, Luchansky M, Figer A, Shapira J, Gottfried M, Novis B, Bogelman G, Zemer R, Zimlichman S, Klein A. Expression of cytokeratin 20 in the blood of patients with disseminated carcinoma of the pancreas, colon, stomach, and lung. Cancer. 1999;86:2398-2405.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Lukyanchuk VV, Friess H, Kleeff J, Osinsky SP, Ayuni E, Candinas D, Roggo A. Detection of circulating tumor cells by cytokeratin 20 and prostate stem cell antigen RT-PCR in blood of patients with gastrointestinal cancers. Anticancer Res. 2003;23:2711-2716.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Zhang YL, Feng JG, Gou JM, Zhou LX, Wang P. Detection of CK20mRNA in peripheral blood of pancreatic cancer and its clinical significance. World J Gastroenterol. 2005;11:1023-1027.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Soeth E, Grigoleit U, Moellmann B, Röder C, Schniewind B, Kremer B, Kalthoff H, Vogel I. Detection of tumor cell dissemination in pancreatic ductal carcinoma patients by CK 20 RT-PCR indicates poor survival. J Cancer Res Clin Oncol. 2005;131:669-676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 77]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
37.  Clarke LE, Leitzel K, Smith J, Ali SM, Lipton A. Epidermal growth factor receptor mRNA in peripheral blood of patients with pancreatic, lung, and colon carcinomas detected by RT-PCR. Int J Oncol. 2003;22:425-430.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Ishizone S, Yamauchi K, Kawa S, Suzuki T, Shimizu F, Harada O, Sugiyama A, Miyagawa S, Fukuda M, Nakayama J. Clinical utility of quantitative RT-PCR targeted to alpha1,4-N-acetylglucosaminyltransferase mRNA for detection of pancreatic cancer. Cancer Sci. 2006;97:119-126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 22]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
39.  Hoffmann K, Kerner C, Wilfert W, Mueller M, Thiery J, Hauss J, Witzigmann H. Detection of disseminated pancreatic cells by amplification of cytokeratin-19 with quantitative RT-PCR in blood, bone marrow and peritoneal lavage of pancreatic carcinoma patients. World J Gastroenterol. 2007;13:257-263.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Zhou J, Hu L, Yu Z, Zheng J, Yang D, Bouvet M, Hoffman RM. Marker expression in circulating cancer cells of pancreatic cancer patients. J Surg Res. 2011;171:631-636.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 45]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
41.  Kurihara T, Itoi T, Sofuni A, Itokawa F, Tsuchiya T, Tsuji S, Ishii K, Ikeuchi N, Tsuchida A, Kasuya K. Detection of circulating tumor cells in patients with pancreatic cancer: a preliminary result. J Hepatobiliary Pancreat Surg. 2008;15:189-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 118]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
42.  Bidard FC, Huguet F, Louvet C, Mineur L, Bouché O, Chibaudel B, Artru P, Desseigne F, Bachet JB, Mathiot C. Circulating tumor cells in locally advanced pancreatic adenocarcinoma: the ancillary CirCe 07 study to the LAP 07 trial. Ann Oncol. 2013;24:2057-2061.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 141]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
43.  Iwanicki-Caron I, Basile P, Toure E, Antonietti M, Lecleire S, Di Fiore A, Oden-Gangloff A, Blanchard F, Lemoine F, Di Fiore F. Usefulness of circulating tumor cell detection in pancreatic adenocarcinoma diagnosis. Am J Gastroenterol. 2013;108:152-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
44.  Cauley CE, Pitman MB, Zhou J, Perkins J, Kuleman B, Liss AS, Fernandez-Del Castillo C, Warshaw AL, Lillemoe KD, Thayer SP. Circulating Epithelial Cells in Patients with Pancreatic Lesions: Clinical and Pathologic Findings. J Am Coll Surg. 2015;221:699-707.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 49]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
45.  Bobek V, Gurlich R, Eliasova P, Kolostova K. Circulating tumor cells in pancreatic cancer patients: enrichment and cultivation. World J Gastroenterol. 2014;20:17163-17170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 57]  [Cited by in F6Publishing: 60]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
46.  Kulemann B, Pitman MB, Liss AS, Valsangkar N, Fernández-Del Castillo C, Lillemoe KD, Hoeppner J, Mino-Kenudson M, Warshaw AL, Thayer SP. Circulating tumor cells found in patients with localized and advanced pancreatic cancer. Pancreas. 2015;44:547-550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 74]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
47.  Rhim AD, Thege FI, Santana SM, Lannin TB, Saha TN, Tsai S, Maggs LR, Kochman ML, Ginsberg GG, Lieb JG. Detection of circulating pancreas epithelial cells in patients with pancreatic cystic lesions. Gastroenterology. 2014;146:647-651.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 173]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
48.  Sheng W, Ogunwobi OO, Chen T, Zhang J, George TJ, Liu C, Fan ZH. Capture, release and culture of circulating tumor cells from pancreatic cancer patients using an enhanced mixing chip. Lab Chip. 2014;14:89-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 198]  [Cited by in F6Publishing: 148]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
49.  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: ]
50.  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: ]
51.  Vasioukhin V, Anker P, Maurice P, Lyautey J, Lederrey C, Stroun M. Point mutations of the N-ras gene in the blood plasma DNA of patients with myelodysplastic syndrome or acute myelogenous leukaemia. Br J Haematol. 1994;86:774-779.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Cachia PG, Taylor C, Thompson PW, Tennant GB, Masters G, Pettersson T, Whittaker JA, Burnett AK, Jacobs A, Padua RA. Non-dysplastic myelodysplasia? Leukemia. 1994;8:677-681.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  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: ]
54.  Kopreski MS, Benko FA, Kwak LW, Gocke CD. Detection of tumor messenger RNA in the serum of patients with malignant melanoma. Clin Cancer Res. 1999;5:1961-1965.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Lo KW, Lo YM, Leung SF, Tsang YS, Chan LY, Johnson PJ, Hjelm NM, Lee JC, Huang DP. Analysis of cell-free Epstein-Barr virus associated RNA in the plasma of patients with nasopharyngeal carcinoma. Clin Chem. 1999;45:1292-1294.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Dasí F, Martínez-Rodes P, March JA, Santamaría J, Martínez-Javaloyas JM, Gil M, Aliño SF. Real-time quantification of human telomerase reverse transcriptase mRNA in the plasma of patients with prostate cancer. Ann N Y Acad Sci. 2006;1075:204-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 36]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
57.  Chen XQ, Bonnefoi H, Pelte MF, Lyautey J, Lederrey C, Movarekhi S, Schaeffer P, Mulcahy HE, Meyer P, Stroun M. Telomerase RNA as a detection marker in the serum of breast cancer patients. Clin Cancer Res. 2000;6:3823-3826.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Silva JM, Rodriguez R, Garcia JM, Muñoz C, Silva J, Dominguez G, Provencio M, España P, Bonilla F. Detection of epithelial tumour RNA in the plasma of colon cancer patients is associated with advanced stages and circulating tumour cells. Gut. 2002;50:530-534.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O’Briant KC, Allen A. 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)]
60.  Diehl F, Li M, Dressman D, He Y, Shen D, Szabo S, Diaz LA, Goodman SN, David KA, Juhl H. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc Natl Acad Sci USA. 2005;102:16368-16373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 840]  [Cited by in F6Publishing: 898]  [Article Influence: 47.3]  [Reference Citation Analysis (0)]
61.  van 't Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, Peterse HL, van der Kooy K, Marton MJ, Witteveen AT. Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002;415:530-536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6904]  [Cited by in F6Publishing: 6230]  [Article Influence: 283.2]  [Reference Citation Analysis (0)]
62.  Dawson SJ, Tsui DW, Murtaza M, Biggs H, Rueda OM, Chin SF, Dunning MJ, Gale D, Forshew T, Mahler-Araujo B. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013;368:1199-1209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1713]  [Cited by in F6Publishing: 1600]  [Article Influence: 145.5]  [Reference Citation Analysis (0)]
63.  Chan KC, Jiang P, Zheng YW, Liao GJ, Sun H, Wong J, Siu SS, Chan WC, Chan SL, Chan AT. Cancer genome scanning in plasma: detection of tumor-associated copy number aberrations, single-nucleotide variants, and tumoral heterogeneity by massively parallel sequencing. Clin Chem. 2013;59:211-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 369]  [Cited by in F6Publishing: 395]  [Article Influence: 35.9]  [Reference Citation Analysis (0)]
64.  Shapiro B, Chakrabarty M, Cohn EM, Leon SA. Determination of circulating DNA levels in patients with benign or malignant gastrointestinal disease. Cancer. 1983;51:2116-2120.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Melnikov AA, Scholtens D, Talamonti MS, Bentrem DJ, Levenson VV. Methylation profile of circulating plasma DNA in patients with pancreatic cancer. J Surg Oncol. 2009;99:119-122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 62]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
66.  Liggett T, Melnikov A, Yi QL, Replogle C, Brand R, Kaul K, Talamonti M, Abrams RA, Levenson V. Differential methylation of cell-free circulating DNA among patients with pancreatic cancer versus chronic pancreatitis. Cancer. 2010;116:1674-1680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 102]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
67.  Moriyama H, Matsubara N, Kanbara T, Mori M, Matsuoka J, Yoshino T, Takakura N, Shimizu K, Takaka N. Allelic imbalance and microsatellite instability in plasma DNA released from polyclonal pancreatic adenocarcinoma. Int J Oncol. 2002;21:949-956.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Smit VT, Boot AJ, Smits AM, Fleuren GJ, Cornelisse CJ, Bos JL. KRAS codon 12 mutations occur very frequently in pancreatic adenocarcinomas. Nucleic Acids Res. 1988;16:7773-7782.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Almoguera C, Shibata D, Forrester K, Martin J, Arnheim N, Perucho M. Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell. 1988;53:549-554.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Tada M, Omata M, Ohto M. Clinical application of ras gene mutation for diagnosis of pancreatic adenocarcinoma. Gastroenterology. 1991;100:233-238.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Lemoine NR, Jain S, Hughes CM, Staddon SL, Maillet B, Hall PA, Klöppel G. Ki-ras oncogene activation in preinvasive pancreatic cancer. Gastroenterology. 1992;102:230-236.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Yamada T, Nakamori S, Ohzato H, Oshima S, Aoki T, Higaki N, Sugimoto K, Akagi K, Fujiwara Y, Nishisho I. Detection of K-ras gene mutations in plasma DNA of patients with pancreatic adenocarcinoma: correlation with clinicopathological features. Clin Cancer Res. 1998;4:1527-1532.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Theodor L, Melzer E, Sologov M, Idelman G, Friedman E, Bar-Meir S. Detection of pancreatic carcinoma: diagnostic value of K-ras mutations in circulating DNA from serum. Dig Dis Sci. 1999;44:2014-2019.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Castells A, Puig P, Móra J, Boadas J, Boix L, Urgell E, Solé M, Capellà G, Lluís F, Fernández-Cruz L. K-ras mutations in DNA extracted from the plasma of patients with pancreatic carcinoma: diagnostic utility and prognostic significance. J Clin Oncol. 1999;17:578-584.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Zambon C, Navaglia F, Basso D, Gallo N, Greco E, Piva MG, Fogar P, Pasquali C, Pedrazzoli S, Plebani M. ME-PCR for the identification of mutated K-ras in serum and bile of pancreatic cancer patients: an unsatisfactory technique for clinical applications. Clin Chim Acta. 2000;302:35-48.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Maire F, Micard S, Hammel P, Voitot H, Lévy P, Cugnenc PH, Ruszniewski P, Puig PL. Differential diagnosis between chronic pancreatitis and pancreatic cancer: value of the detection of KRAS2 mutations in circulating DNA. Br J Cancer. 2002;87:551-554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 109]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
77.  Marchese R, Muleti A, Brozzetti S, Gandini O, Brunetti E, French D. Low value of detection of KRAS2 mutations in circulating DNA to differentiate chronic pancreatitis to pancreatic cancer. Br J Cancer. 2004;90:2243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
78.  Magistrelli P, Neri M, Granone P, Cesario A, Paleari L, Russo P. K-ras mutations in circulating DNA from pancreatic and lung cancers: bridging methodology for a common validation of the molecular diagnosis value. Pancreas. 2008;37:101-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
79.  Chen H, Tu H, Meng ZQ, Chen Z, Wang P, Liu LM. K-ras mutational status predicts poor prognosis in unresectable pancreatic cancer. Eur J Surg Oncol. 2010;36:657-662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 97]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
80.  Wu J, Zhou Y, Zhang CY, Song BB, Wang BL, Pan BS, Lou WH, Guo W. Co-amplification at lower denaturation-temperature PCR combined with unlabled-probe high-resolution melting to detect KRAS codon 12 and 13 mutations in plasma-circulating DNA of pancreatic adenocarcinoma cases. Asian Pac J Cancer Prev. 2014;15:10647-10652.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Earl J, Garcia-Nieto S, Martinez-Avila JC, Montans J, Sanjuanbenito A, Rodríguez-Garrote M, Lisa E, Mendía E, Lobo E, Malats N. Circulating tumor cells (Ctc) and kras mutant circulating free Dna (cfdna) detection in peripheral blood as biomarkers in patients diagnosed with exocrine pancreatic cancer. BMC Cancer. 2015;15:797.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 132]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
82.  Kinugasa H, Nouso K, Miyahara K, Morimoto Y, Dohi C, Tsutsumi K, Kato H, Matsubara T, Okada H, Yamamoto K. Detection of K-ras gene mutation by liquid biopsy in patients with pancreatic cancer. Cancer. 2015;121:2271-2280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 171]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
83.  Sausen M, Phallen J, Adleff V, Jones S, Leary RJ, Barrett MT, Anagnostou V, Parpart-Li S, Murphy D, Kay Li Q. Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients. Nat Commun. 2015;6:7686.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 295]  [Cited by in F6Publishing: 322]  [Article Influence: 35.8]  [Reference Citation Analysis (0)]
84.  Takai E, Totoki Y, Nakamura H, Morizane C, Nara S, Hama N, Suzuki M, Furukawa E, Kato M, Hayashi H. Clinical utility of circulating tumor DNA for molecular assessment in pancreatic cancer. Sci Rep. 2015;5:18425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 134]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
85.  Zill OA, Greene C, Sebisanovic D, Siew LM, Leng J, Vu M, Hendifar AE, Wang Z, Atreya CE, Kelley RK. Cell-Free DNA Next-Generation Sequencing in Pancreatobiliary Carcinomas. Cancer Discov. 2015;5:1040-1048.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 187]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
86.  Diaz LA, Williams RT, Wu J, Kinde I, Hecht JR, Berlin J, Allen B, Bozic I, Reiter JG, Nowak MA. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature. 2012;486:537-540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1259]  [Cited by in F6Publishing: 1292]  [Article Influence: 107.7]  [Reference Citation Analysis (0)]
87.  Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, Valtorta E, Schiavo R, Buscarino M, Siravegna G. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 2012;486:532-536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1313]  [Cited by in F6Publishing: 1373]  [Article Influence: 114.4]  [Reference Citation Analysis (0)]
88.  Murtaza M, Dawson SJ, Tsui DW, Gale D, Forshew T, Piskorz AM, Parkinson C, Chin SF, Kingsbury Z, Wong AS. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497:108-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1288]  [Cited by in F6Publishing: 1252]  [Article Influence: 113.8]  [Reference Citation Analysis (0)]
89.  Reddi KK, Holland JF. Elevated serum ribonuclease in patients with pancreatic cancer. Proc Natl Acad Sci USA. 1976;73:2308-2310.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Houseley J, LaCava J, Tollervey D. RNA-quality control by the exosome. Nat Rev Mol Cell Biol. 2006;7:529-539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 503]  [Cited by in F6Publishing: 515]  [Article Influence: 28.6]  [Reference Citation Analysis (0)]
91.  Silva JM, Dominguez G, Silva J, Garcia JM, Sanchez A, Rodriguez O, Provencio M, España P, Bonilla F. Detection of epithelial messenger RNA in the plasma of breast cancer patients is associated with poor prognosis tumor characteristics. Clin Cancer Res. 2001;7:2821-2825.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Wong SC, Lo SF, Cheung MT, Ng KO, Tse CW, Lai BS, Lee KC, Lo YM. Quantification of plasma beta-catenin mRNA in colorectal cancer and adenoma patients. Clin Cancer Res. 2004;10:1613-1617.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Garcia V, García JM, Peña C, Silva J, Domínguez G, Hurtado A, Alonso I, Rodriguez R, Provencio M, Bonilla F. Thymidylate synthase messenger RNA expression in plasma from patients with colon cancer: prognostic potential. Clin Cancer Res. 2006;12:2095-2100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 32]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
94.  Kang CY, Wang J, Axell-House D, Soni P, Chu ML, Chipitsyna G, Sarosiek K, Sendecki J, Hyslop T, Al-Zoubi M. Clinical significance of serum COL6A3 in pancreatic ductal adenocarcinoma. J Gastrointest Surg. 2014;18:7-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
95.  ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14285]  [Cited by in F6Publishing: 11818]  [Article Influence: 984.8]  [Reference Citation Analysis (0)]
96.  Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W. Initial sequencing and analysis of the human genome. Nature. 2001;409:860-921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16054]  [Cited by in F6Publishing: 14483]  [Article Influence: 629.7]  [Reference Citation Analysis (0)]
97.  Venter JC, Adams MD, Myers EW, Li PW, Mural RJ, Sutton GG, Smith HO, Yandell M, Evans CA, Holt RA. The sequence of the human genome. Science. 2001;291:1304-1351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9175]  [Cited by in F6Publishing: 7670]  [Article Influence: 333.5]  [Reference Citation Analysis (0)]
98.  Szymanski M, Barciszewska MZ, Erdmann VA, Barciszewski J. A new frontier for molecular medicine: noncoding RNAs. Biochim Biophys Acta. 2005;1756:65-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 46]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
99.  Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654-659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8246]  [Cited by in F6Publishing: 9027]  [Article Influence: 531.0]  [Reference Citation Analysis (0)]
100.  Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, Matsuki Y, Ochiya T. Secretory mechanisms and intercellular transfer of microRNAs in living cells. J Biol Chem. 2010;285:17442-17452.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1382]  [Cited by in F6Publishing: 1491]  [Article Influence: 106.5]  [Reference Citation Analysis (0)]
101.  Arroyo JD, Chevillet JR, Kroh EM, Ruf IK, Pritchard CC, Gibson DF, Mitchell PS, Bennett CF, Pogosova-Agadjanyan EL, Stirewalt DL. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci USA. 2011;108:5003-5008.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2345]  [Cited by in F6Publishing: 2488]  [Article Influence: 191.4]  [Reference Citation Analysis (0)]
102.  Wang J, Chen J, Chang P, LeBlanc A, Li D, Abbruzzesse JL, Frazier ML, Killary AM, Sen S. MicroRNAs in plasma of pancreatic ductal adenocarcinoma patients as novel blood-based biomarkers of disease. Cancer Prev Res (Phila). 2009;2:807-813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 392]  [Cited by in F6Publishing: 418]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
103.  Schultz NA, Dehlendorff C, Jensen BV, Bjerregaard JK, Nielsen KR, Bojesen SE, Calatayud D, Nielsen SE, Yilmaz M, Holländer NH. MicroRNA biomarkers in whole blood for detection of pancreatic cancer. JAMA. 2014;311:392-404.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 317]  [Cited by in F6Publishing: 321]  [Article Influence: 32.1]  [Reference Citation Analysis (0)]
104.  Kojima M, Sudo H, Kawauchi J, Takizawa S, Kondou S, Nobumasa H, Ochiai A. MicroRNA markers for the diagnosis of pancreatic and biliary-tract cancers. PLoS One. 2015;10:e0118220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 96]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
105.  Kong X, Du Y, Wang G, Gao J, Gong Y, Li L, Zhang Z, Zhu J, Jing Q, Qin Y. Detection of differentially expressed microRNAs in serum of pancreatic ductal adenocarcinoma patients: miR-196a could be a potential marker for poor prognosis. Dig Dis Sci. 2011;56:602-609.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 122]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
106.  Miyamae M, Komatsu S, Ichikawa D, Kawaguchi T, Hirajima S, Okajima W, Ohashi T, Imamura T, Konishi H, Shiozaki A. Plasma microRNA profiles: identification of miR-744 as a novel diagnostic and prognostic biomarker in pancreatic cancer. Br J Cancer. 2015;113:1467-1476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 79]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
107.  Kim K, Jutooru I, Chadalapaka G, Johnson G, Frank J, Burghardt R, Kim S, Safe S. HOTAIR is a negative prognostic factor and exhibits pro-oncogenic activity in pancreatic cancer. Oncogene. 2013;32:1616-1625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 578]  [Cited by in F6Publishing: 658]  [Article Influence: 54.8]  [Reference Citation Analysis (0)]
108.  Pang EJ, Yang R, Fu XB, Liu YF. Overexpression of long non-coding RNA MALAT1 is correlated with clinical progression and unfavorable prognosis in pancreatic cancer. Tumour Biol. 2015;36:2403-2407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 149]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
109.  Modali SD, Parekh VI, Kebebew E, Agarwal SK. Epigenetic regulation of the lncRNA MEG3 and its target c-MET in pancreatic neuroendocrine tumors. Mol Endocrinol. 2015;29:224-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 90]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
110.  Lu X, Fang Y, Wang Z, Xie J, Zhan Q, Deng X, Chen H, Jin J, Peng C, Li H. Downregulation of gas5 increases pancreatic cancer cell proliferation by regulating CDK6. Cell Tissue Res. 2013;354:891-896.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 124]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
111.  Peng W, Gao W, Feng J. Long noncoding RNA HULC is a novel biomarker of poor prognosis in patients with pancreatic cancer. Med Oncol. 2014;31:346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 127]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
112.  Huang C, Yu W, Wang Q, Cui H, Wang Y, Zhang L, Han F, Huang T. Increased expression of the lncRNA PVT1 is associated with poor prognosis in pancreatic cancer patients. Minerva Med. 2015;106:143-149.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Tahira AC, Kubrusly MS, Faria MF, Dazzani B, Fonseca RS, Maracaja-Coutinho V, Verjovski-Almeida S, Machado MC, Reis EM. Long noncoding intronic RNAs are differentially expressed in primary and metastatic pancreatic cancer. Mol Cancer. 2011;10:141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 135]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
114.  Li J, Liu D, Hua R, Zhang J, Liu W, Huo Y, Cheng Y, Hong J, Sun Y. Long non-coding RNAs expressed in pancreatic ductal adenocarcinoma and lncRNA BC008363 an independent prognostic factor in PDAC. Pancreatology. 2014;14:385-390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
115.  Sun YW, Chen YF, Li J, Huo YM, Liu DJ, Hua R, Zhang JF, Liu W, Yang JY, Fu XL. A novel long non-coding RNA ENST00000480739 suppresses tumour cell invasion by regulating OS-9 and HIF-1α in pancreatic ductal adenocarcinoma. Br J Cancer. 2014;111:2131-2141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 82]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
116.  Ting DT, Lipson D, Paul S, Brannigan BW, Akhavanfard S, Coffman EJ, Contino G, Deshpande V, Iafrate AJ, Letovsky S. Aberrant overexpression of satellite repeats in pancreatic and other epithelial cancers. Science. 2011;331:593-596.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 364]  [Cited by in F6Publishing: 386]  [Article Influence: 29.7]  [Reference Citation Analysis (0)]
117.  Wang Y, Li Z, Zheng S, Zhou Y, Zhao L, Ye H, Zhao X, Gao W, Fu Z, Zhou Q. Expression profile of long non-coding RNAs in pancreatic cancer and their clinical significance as biomarkers. Oncotarget. 2015;6:35684-35698.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 75]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
118.  Záveský L, Jandáková E, Turyna R, Langmeierová L, Weinberger V, Záveská Drábková L, Hůlková M, Hořínek A, Dušková D, Feyereisl J. Evaluation of Cell-Free Urine microRNAs Expression for the Use in Diagnosis of Ovarian and Endometrial Cancers. A Pilot Study. Pathol Oncol Res. 2015;21:1027-1035.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 37]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
119.  Erbes T, Hirschfeld M, Rücker G, Jaeger M, Boas J, Iborra S, Mayer S, Gitsch G, Stickeler E. Feasibility of urinary microRNA detection in breast cancer patients and its potential as an innovative non-invasive biomarker. BMC Cancer. 2015;15:193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 97]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
120.  Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, Kamiyama M, Hruban RH, Eshleman JR, Nowak MA. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467:1114-1117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1872]  [Cited by in F6Publishing: 1837]  [Article Influence: 131.2]  [Reference Citation Analysis (0)]
121.  Cooper CL, O’Toole SA, Kench JG. Classification, morphology and molecular pathology of premalignant lesions of the pancreas. Pathology. 2013;45:286-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
122.  Permuth-Wey J, Chen DT, Fulp WJ, Yoder SJ, Zhang Y, Georgeades C, Husain K, Centeno BA, Magliocco AM, Coppola D. Plasma MicroRNAs as Novel Biomarkers for Patients with Intraductal Papillary Mucinous Neoplasms of the Pancreas. Cancer Prev Res (Phila). 2015;8:826-834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 42]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
123.  Komatsu S, Ichikawa D, Miyamae M, Kawaguchi T, Morimura R, Hirajima S, Okajima W, Ohashi T, Imamura T, Konishi H. Malignant potential in pancreatic neoplasm; new insights provided by circulating miR-223 in plasma. Expert Opin Biol Ther. 2015;15:773-785.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 46]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
124.  Z'graggen K, Centeno BA, Fernandez-del Castillo C, Jimenez RE, Werner J, Warshaw AL. Biological implications of tumor cells in blood and bone marrow of pancreatic cancer patients. Surgery. 2001;129:537-546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 76]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
125.  Khoja L, Backen A, Sloane R, Menasce L, Ryder D, Krebs M, Board R, Clack G, Hughes A, Blackhall F. A pilot study to explore circulating tumour cells in pancreatic cancer as a novel biomarker. Br J Cancer. 2012;106:508-516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 201]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
126.  de Albuquerque A, Kubisch I, Breier G, Stamminger G, Fersis N, Eichler A, Kaul S, Stölzel U. Multimarker gene analysis of circulating tumor cells in pancreatic cancer patients: a feasibility study. Oncology. 2012;82:3-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 89]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
127.  Kamande JW, Hupert ML, Witek MA, Wang H, Torphy RJ, Dharmasiri U, Njoroge SK, Jackson JM, Aufforth RD, Snavely A. Modular microsystem for the isolation, enumeration, and phenotyping of circulating tumor cells in patients with pancreatic cancer. Anal Chem. 2013;85:9092-9100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 89]  [Article Influence: 8.1]  [Reference Citation Analysis (1)]
128.  Catenacci DV, Chapman CG, Xu P, Koons A, Konda VJ, Siddiqui UD, Waxman I. Acquisition of Portal Venous Circulating Tumor Cells From Patients With Pancreaticobiliary Cancers by Endoscopic Ultrasound. Gastroenterology. 2015;149:1794-1803.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 113]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
129.  Zhang Y, Wang F, Ning N, Chen Q, Yang Z, Guo Y, Xu D, Zhang D, Zhan T, Cui W. Patterns of circulating tumor cells identified by CEP8, CK and CD45 in pancreatic cancer. Int J Cancer. 2015;136:1228-1233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 71]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
130.  Bissolati M, Sandri MT, Burtulo G, Zorzino L, Balzano G, Braga M. Portal vein-circulating tumor cells predict liver metastases in patients with resectable pancreatic cancer. Tumour Biol. 2015;36:991-996.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 78]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
131.  Zhang J, Li S, Liu F, Zhou L, Shao N, Zhao X. SELEX aptamer used as a probe to detect circulating tumor cells in peripheral blood of pancreatic cancer patients. PLoS One. 2015;10:e0121920.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
132.  Giacona MB, Ruben GC, Iczkowski KA, Roos TB, Porter DM, Sorenson GD. Cell-free DNA in human blood plasma: length measurements in patients with pancreatic cancer and healthy controls. Pancreas. 1998;17:89-97.  [PubMed]  [DOI]  [Cited in This Article: ]
133.  Singh N, Gupta S, Pandey RM, Chauhan SS, Saraya A. High levels of cell-free circulating nucleic acids in pancreatic cancer are associated with vascular encasement, metastasis and poor survival. Cancer Invest. 2015;33:78-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 68]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
134.  Ho AS, Huang X, Cao H, Christman-Skieller C, Bennewith K, Le QT, Koong AC. Circulating miR-210 as a Novel Hypoxia Marker in Pancreatic Cancer. Transl Oncol. 2010;3:109-113.  [PubMed]  [DOI]  [Cited in This Article: ]
135.  Li A, Omura N, Hong SM, Vincent A, Walter K, Griffith M, Borges M, Goggins M. Pancreatic cancers epigenetically silence SIP1 and hypomethylate and overexpress miR-200a/200b in association with elevated circulating miR-200a and miR-200b levels. Cancer Res. 2010;70:5226-5237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 245]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
136.  Ali S, Almhanna K, Chen W, Philip PA, Sarkar FH. Differentially expressed miRNAs in the plasma may provide a molecular signature for aggressive pancreatic cancer. Am J Transl Res. 2010;3:28-47.  [PubMed]  [DOI]  [Cited in This Article: ]
137.  LaConti JJ, Shivapurkar N, Preet A, Deslattes Mays A, Peran I, Kim SE, Marshall JL, Riegel AT, Wellstein A. Tissue and serum microRNAs in the Kras(G12D) transgenic animal model and in patients with pancreatic cancer. PLoS One. 2011;6:e20687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 75]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
138.  Morimura R, Komatsu S, Ichikawa D, Takeshita H, Tsujiura M, Nagata H, Konishi H, Shiozaki A, Ikoma H, Okamoto K. Novel diagnostic value of circulating miR-18a in plasma of patients with pancreatic cancer. Br J Cancer. 2011;105:1733-1740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 202]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
139.  Liu J, Gao J, Du Y, Li Z, Ren Y, Gu J, Wang X, Gong Y, Wang W, Kong X. Combination of plasma microRNAs with serum CA19-9 for early detection of pancreatic cancer. Int J Cancer. 2012;131:683-691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 210]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
140.  Liu R, Chen X, Du Y, Yao W, Shen L, Wang C, Hu Z, Zhuang R, Ning G, Zhang C. Serum microRNA expression profile as a biomarker in the diagnosis and prognosis of pancreatic cancer. Clin Chem. 2012;58:610-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 276]  [Cited by in F6Publishing: 294]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
141.  Li A, Yu J, Kim H, Wolfgang CL, Canto MI, Hruban RH, Goggins M. MicroRNA array analysis finds elevated serum miR-1290 accurately distinguishes patients with low-stage pancreatic cancer from healthy and disease controls. Clin Cancer Res. 2013;19:3600-3610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 225]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
142.  Wang WS, Liu LX, Li GP, Chen Y, Li CY, Jin DY, Wang XL. Combined serum CA19-9 and miR-27a-3p in peripheral blood mononuclear cells to diagnose pancreatic cancer. Cancer Prev Res (Phila). 2013;6:331-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 65]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
143.  Kawaguchi T, Komatsu S, Ichikawa D, Morimura R, Tsujiura M, Konishi H, Takeshita H, Nagata H, Arita T, Hirajima S. Clinical impact of circulating miR-221 in plasma of patients with pancreatic cancer. Br J Cancer. 2013;108:361-369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 175]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
144.  Zhao C, Zhang J, Zhang S, Yu D, Chen Y, Liu Q, Shi M, Ni C, Zhu M. Diagnostic and biological significance of microRNA-192 in pancreatic ductal adenocarcinoma. Oncol Rep. 2013;30:276-284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 76]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
145.  Wang P, Zhuang L, Zhang J, Fan J, Luo J, Chen H, Wang K, Liu L, Chen Z, Meng Z. The serum miR-21 level serves as a predictor for the chemosensitivity of advanced pancreatic cancer, and miR-21 expression confers chemoresistance by targeting FasL. Mol Oncol. 2013;7:334-345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 150]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
146.  Carlsen AL, Joergensen MT, Knudsen S, de Muckadell OB, Heegaard NH. Cell-free plasma microRNA in pancreatic ductal adenocarcinoma and disease controls. Pancreas. 2013;42:1107-1113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
147.  Que R, Ding G, Chen J, Cao L. Analysis of serum exosomal microRNAs and clinicopathologic features of patients with pancreatic adenocarcinoma. World J Surg Oncol. 2013;11:219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 198]  [Article Influence: 18.0]  [Reference Citation Analysis (1)]
148.  Gao L, He SB, Li DC. Effects of miR-16 plus CA19-9 detections on pancreatic cancer diagnostic performance. Clin Lab. 2014;60:73-77.  [PubMed]  [DOI]  [Cited in This Article: ]
149.  Chen Q, Yang L, Xiao Y, Zhu J, Li Z. Circulating microRNA-182 in plasma and its potential diagnostic and prognostic value for pancreatic cancer. Med Oncol. 2014;31:225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 38]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
150.  Ganepola GA, Rutledge JR, Suman P, Yiengpruksawan A, Chang DH. Novel blood-based microRNA biomarker panel for early diagnosis of pancreatic cancer. World J Gastrointest Oncol. 2014;6:22-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 88]  [Cited by in F6Publishing: 81]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
151.  Abue M, Yokoyama M, Shibuya R, Tamai K, Yamaguchi K, Sato I, Tanaka N, Hamada S, Shimosegawa T, Sugamura K. Circulating miR-483-3p and miR-21 is highly expressed in plasma of pancreatic cancer. Int J Oncol. 2015;46:539-547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 140]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
152.  Slater EP, Strauch K, Rospleszcz S, Ramaswamy A, Esposito I, Klöppel G, Matthäi E, Heeger K, Fendrich V, Langer P. MicroRNA-196a and -196b as Potential Biomarkers for the Early Detection of Familial Pancreatic Cancer. Transl Oncol. 2014;7:464-471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 58]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
153.  Xu J, Cao Z, Liu W, You L, Zhou L, Wang C, Lou W, Sun B, Miao Y, Liu X. Plasma miRNAs Effectively Distinguish Patients With Pancreatic Cancer From Controls: A Multicenter Study. Ann Surg. 2016;263:1173-1179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 64]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
154.  Madhavan B, Yue S, Galli U, Rana S, Gross W, Müller M, Giese NA, Kalthoff H, Becker T, Büchler MW. Combined evaluation of a panel of protein and miRNA serum-exosome biomarkers for pancreatic cancer diagnosis increases sensitivity and specificity. Int J Cancer. 2015;136:2616-2627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 314]  [Cited by in F6Publishing: 351]  [Article Influence: 35.1]  [Reference Citation Analysis (0)]
155.  Baraniskin A, Nöpel-Dünnebacke S, Ahrens M, Jensen SG, Zöllner H, Maghnouj A, Wos A, Mayerle J, Munding J, Kost D. Circulating U2 small nuclear RNA fragments as a novel diagnostic biomarker for pancreatic and colorectal adenocarcinoma. Int J Cancer. 2013;132:E48-E57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 110]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]