Abstract
Pancreatic cancer (PC) is characterized by extremely high mortality and poor prognosis, which are largely ascribed to difficulties in early diagnosis and limited therapeutics. Although there is a sufficient window for intervention before preneoplastic lesions progress to invasive disease, effective early detection of PC remains difficult using current biomarkers and imaging techniques. Biomarkers with satisfactory diagnostic efficacy and convenient analysis methods are urgently required. In this review, we summarized recent advances in the identification of biomarkers in circulation for early detection of PC. A number of novel circulating biomarkers, such as metabolites, cell-free DNA (cfDNA), noncoding RNA, and exosomes, that show promising diagnostic value have been discovered using advances in sequencing techniques and “omics” analyses. Panels comprising several biomarkers may also exhibit better diagnostic performance. In the future, we need more efficient circulating biomarkers for the identification of noninvasive precursor lesions and early disease. Collaborative large-scale studies are also required to show the clinical validity and applicability of potential biomarkers.
Keywords: Pancreatic cancer, early diagnosis, circulating biomarkers
Introduction
Pancreatic cancer (PC) is characterized by extremely high mortality and poor prognosis and is the 4th leading cause of cancer-related death [1]. Unfortunately, compared with other malignancies, there has been little improvement in the survival rate of patients with PC in recent decades.
Surgical resection is currently the only therapeutic that can cure early-stage PC. However, because PC patients mostly present with no clinically informative symptoms or radiologic manifestations at early stages [2], the majority (approximately 80%) of PC patients are diagnosed with locally advanced or metastatic disease and do not have the opportunity to undergo a curative operation [3]. The 5-year survival rate would increase significantly if PC patients were identified at an earlier stage and underwent surgical resection followed by chemotherapy [4-6]. Therefore, the most promising direction for improving the prognosis of PC is to develop effective screening strategies with which to detect the disease at an earlier stage. There is a possible window of opportunity for the detection of PC from the initial mutations in the pancreas to the development of metastatic sites, suggesting that early detection is feasible in this period, which can span two decades [7].
However, early diagnosis of PC is difficult with the currently available methods [8]. Unlike colonoscopies for colorectal cancer and serum prostate-specific antigen (PSA) levels for prostate cancer, there is currently no standardized PC screening strategy, even for high-risk populations. The diagnosis of PC is usually based on radiology [computed tomography (CT) and magnetic resonance imaging (MRI)] or invasive endoscopic techniques [ultrasound endoscopy-fine needle aspiration (EUS-FNA), endoscopic retrograde cholangiopancreatography (ERCP), and explorative laparoscopy] [9], which are inconvenient for patients and show no superiority over biological markers. Furthermore, PC is characterized by dense desmoplasia surrounded by a large amount of extracellular matrix, creating a hurdle for pathologic biopsy [10].
Effective biomarkers that can be obtained in a less invasive manner have become a research focus. The ideal biomarkers should be easily detected with satisfactory sensitivity and specificity and should distinguish PC from other benign pancreatic lesions. In the context of early detection, the identification of preneoplastic conditions, such as pancreatic intraepithelial neoplasia (PanIN), intraductal papillary mucinous neoplasms (IPMNs) and mucinous cystic neoplasms (MCNs), is of great importance [11].
Blood is easily accessible and relatively stable, making serum an ideal specimen in which to discover biomarkers. However, biomarkers secreted into serum are extremely dilute and probably obscured by other more-abundant serum proteins [12]. Technological advances in the last decade have provided more opportunities to discover circulating biomarkers based on “omics” analyses, including methods focused on proteins, nucleic acids, circulating tumor cells (CTCs), and exosomes. Numerous proteins of low abundance can be analyzed by mass spectrometry-based approaches and proteomic technologies. Next-generation sequencing techniques provide deeper insight into somatic mutations and epigenetics analysis of the genome and broaden the characterization of circulating tumor DNA (ctDNA) and cell-free RNA. With the development of cell tracking techniques and flow cytometry, it is now possible to capture and analyze CTCs and exosomes.
Thus, in this review, we summarize recent progress in the early detection of PC using various types of circulating biomarkers (Figure 1).
CA19-9, other carbohydrate antigens, and carcinoembryonic antigen
CA19-9, also called sialyl Lewis a, is the only biomarker approved by the US FDA for monitoring the progression and therapeutic response of PC; it has also been widely used in the diagnosis of PC for a long time [13,14]. However, the reported sensitivity (ranging from 69% to 98%) and specificity (ranging from 46% to 98%) of CA19-9 are moderate for PC screening [15-18].
There are some challenges facing the use of CA19-9 alone as a biomarker for the early detection of PC. CA19-9 is an epitope of sialylated Lewis group antigen, and approximately 5-10% of the Caucasian population possesses a Lewis a-/b- genotype and thus does not express CA19-9 [19]. Furthermore, only 65% of patients with resectable PC have elevated serum CA19-9 levels [20]; therefore, CA19-9 testing would result in false-negative results and mislead the diagnosis. Third, an elevated level of CA19-9 has been observed in other gastrointestinal malignancies, such as gastric cancer and colorectal cancer, and in various benign conditions, including chronic pancreatitis and acute cholangitis [13].
Therefore, recent efforts have been focused on improving the diagnostic efficacy of CA19-9 by combining it with a panel of markers [17,21-23] (Table 1). Makawita et al. [24] combined CA19-9 with SYCN, REG1B and AGR2 to produce a better AUC (AUC=0.87) for diagnosing early-stage PC compared with CA19-9 alone (AUC=0.82, P<0.05). After investigating 83 promising proteins in more than 300 PC patients and controls, Brand et al. [21] discovered a panel consisting of CA19-9, ICAM-1 and OPG that could discriminate PC patients from healthy controls with a sensitivity of 78% and a specificity of 94% in a validation set. Furthermore, this panel is highly selective for PC and yields negative results for almost all breast, lung, and colorectal cancers. Park and colleagues [25] also found a panel comprising CA19-9, cathepsin D and MMP-7 with an AUC of 0.900 for discriminating patients with PC from normal controls.
Table 1.
Classification | Biomarkers in the panel | Participants | Results | Ref. |
---|---|---|---|---|
Concentration measurement | CA19-9, SYCN, REG1B | Early-stage PC vs. NC | AUC=0.87 | Makawita et al. [24] |
CA19-9, ICAM-1, OPG | PC vs. NC | Sen=78%, Spe=94% | Brand et al. [21] | |
CA19-9, cathepsin D, MMP-7 | PC vs. NC | AUC=0.90, Sen=88%, Spe=80% | Park et al. [25] | |
CA19-9, IP-10, IL-6, PDGF | PC vs. non-PC | AUC=0.884 | Shaw et al. [33] | |
HGF, MCP-1, IP-10, Eotaxin | PC vs. NC | Sen=85.7%, Spe=92.3% | Zeh et al. [32] | |
Twenty-five types of inflammatory factors and cytokines | PC vs. NC | AUC=0.95, Sen=88%, Spe=85% | Wingren et al. [34] | |
Metabolomics based on MS | Xylitol, 1,5-anhydro-D-glucitol, histidine, and inositol | PC vs. NC | Sen=84.1%, Spe=84.1% | Kobayashi et al. [48] |
Multi-amino acid-based metabolites | PC vs. NC | AUC=0.891 | Leichtle et al. [53] | |
Methylation status of gene promoters in cfDNA | CCND2, DAPK1, ESR1 promA, HMLH1, MGMT, MUC2, MYOD1, CDKN2B, CDKN1C, PGK1, PGR-proximal, RARb, RB1, SYK | PC vs. CP | Sen=91.2%, Spe=90.8% | Liggett et al. [94] |
CCND2, SOCS1, THBS1, PLAU, VHL | PC vs. NC | Sen=76%, Spe=59% | Melnikov et al. [95] | |
BNC1, ADAMTS1 | PC vs. NC | Sen=81%, Spe=85% | Yi et al. [96] | |
Cell-free ncRNA | Panel 1: CA19-9, miR-145, miR-150, miR-223, miR-636 | PC vs. NC | Panel 1: AUC=0.94, Sen=85%, Spe=85% | Schultz et al. [110] |
Panel 2: CA19-9, miR-26b, miR-34a, miR-122, miR-126*, miR-145, miR-150, miR-223, miR-505, miR-636, miR-885.5p | Panel 2: AUC=0.93, Sen=85%, Spe=85% | |||
Panel 1: miR-486-5p, miR-126-3p, miR-106b-3p | PC vs. CP | Panel 1: AUC=0.891, Sen=82.7%, Spe=84.4% | Cao et al. [111] | |
Panel 2: miR-486-5p, miR-126-3p, miR-106b-3p, miR-938, miR-26b-3p, miR-1285 | Panel 2: AUC=0.889, Sen=82.3%, Spe=81.4% | |||
Biomarkers in circulating exosomes | PC-initiating cell markers (CD44v6, Tspan8, EpCAM, MET, and CD104) and miRNAs (miR-1246, miR-4644, miR-3976, and miR-4306) | PC vs. non-PC | Sen=100%, Spe=80% | Madhavan et al. [172] |
PC: pancreatic cancer; NC: normal controls/healthy controls; CP: chronic pancreatitis; MS: mass spectrometry; AUC: area under curve; Sen: sensitivity; and Spe: specificity.
Other conventional biomarkers, such as CA242, CA72-4, CA125 and carcinoembryonic antigen (CEA), have shown limited diagnostic efficacy for early detection of PC [9,26]; however, in the Lewis-negative population, these biomarkers may have promise. Luo et al. [27] reported that both CEA and CA125 showed high specificity (98.0% and 93.8%, respectively) in Lewis-negative patients with PC and were associated with tumor metastasis and the therapeutic response, which indicates the diagnostic value of elevated CEA and CA125 in patients suspected of having PC.
Additionally, some recent studies have focused on subtypes of carbohydrate antigens. Yue reported that CA19-9 antigenic epitopes on certain mucin backbones, including MUC1, MUC5AC and MUC16, were directly related to the presence of pancreatic pathologies, with better sensitivity and specificity for PC [28]. A study by Partyka et al. [29] reported antibodies with high specificity against Lewis C glycan, a variation of sialyl Lewis a, in diagnosing PC.
In general, panels combining CA19-9 with other novel biomarkers might represent an ideal strategy for improving the sensitivity and specificity of CA19-9 in detecting PC. Insights into the characteristics and mechanisms of CA19-9 and other conventional cancer biomarkers will lead to the optimization of diagnostics using common assays.
Inflammatory factors and growth factors
There is a complicated relationship between inflammation and cancer. Various inflammatory factors, including cytokines, chemokines, and growth factors, have been shown to play important roles in tumorigenesis and metastasis [30], indicating that these factors are potential diagnostic markers of PC.
Growth factor stimulation and neovascularization are critical during the development of carcinomas. Similar to patients with other types of cancer, patients with PC have significantly elevated serum VEGF and bFGF levels compared with healthy individuals [31]. A previous study also reported a correlation between the expression levels of these growth factors and tumor size.
Panels combining multiple cytokines, chemokines, and growth factors were superior to a single biomarker for the detection of PC [32-34]. After measuring serum levels of 27 cytokines in 241 participants, including 127 with PC, Shaw et al. [33] proposed a panel of IP-10, IL-6, PDGF and CA19-9 that offered improved diagnostic performance in the differential diagnosis between PC and benign pancreatic diseases.
Wingren et al. [34] constructed a recombinant antibody microarray to identify different inflammatory factor profiles in serum between PC patients and normal subjects. Some key molecules, such as C1 esterase inhibitor, C3, C5, CD40, CD40 ligand, factor B, GLP-1, IFN-γ, IgM, IL-10, IL-11, IL-12, IL-13, IL-16, IL-18, IL-1-ra, IL-1α, IL-3, IL-5, IL-6, IL-7 and IL-8, integrin-α-11, procathepsin W, sialyl Lewis x, TGF-β1, TNF-α and VEGF, were observed to be differentially overexpressed in a statistically significant manner. Based on these findings, it was concluded that a panel of 25 biomarkers could differentiate patients with PC from normal controls (AUC: 0.95) with a sensitivity and specificity of 88% and 85%, respectively.
Metabolites
Cancer cells are capable of surviving and proliferating in settings with oxygen and nutrient supply deficiency [35] by a process known as the Warburg effect, which was discovered nearly a century ago. Cancer cells are capable of metabolic rewiring, or metabolic reprogramming, to facilitate survival under these conditions. This strategy is extremely important for PC, since PC cells live in a harsh extracellular environment characterized by hypoxia, considerable desmoplasia and hypovascularization [36-38]. Recent technological advances have attracted more attention and interest in cancer-associated metabolic abnormalities and their potential diagnostic and therapeutic applications [39,40]. Accordingly, detection of aberrant low-molecular weight products and intermediates from metabolic reprogramming would indicate an abnormal biochemical state in the individual and suggest the presence of a malignancy. Some groups have reported the application of metabolomic biomarkers in detecting various types of cancer [41,42].
It has been demonstrated that PC cells reprogram intermediary metabolism to meet their metabolic demands, which differ from the demands of normal cells. Until recent years, the distinctive metabolic features of PC were unknown. A large proportion of metabolic rewiring is driven by mutation of the oncogene KRAS, which is almost universally seen in PC cells [43]. To sustain tumor viability, stromal components, including fibroblasts [44,45] and pancreatic stellate cells (PSCs) [46], play supportive roles by mediating metabolite exchange.
Mass spectrometry-based proteomic analysis of PC revealed alterations in metabolic enzymes and the accumulation of key intermediates [47]. Moreover, some amino acids, lipids, fatty acids, and bile acids in serum have been reported as candidate metabolites that may discriminate PC from benign lesions and non-disease states [48-50]. Iole et al. showed that palmitic acid in serum could distinguish PC patients from healthy controls better than traditional CA19-9 [51]. A combination of four serum metabolites (xylitol, 1,5-anhydro-D-glucitol, histidine, and inositol) proposed by Kobayashi et al. had a higher sensitivity in PC and a lower false-negative rate in chronic pancreatitis than CEA and CA19-9 [52]. Leichtle et al. [53] also reported a panel based on serum amino acids for distinguishing PC patients from healthy controls and pancreatitis patients.
Metabolomics is a sensitive indicator for monitoring precancerous lesions (e.g., PanIN) and identifying early-stage PC, as dramatic changes can be observed during different stages of PC progression [54]. Wen et al. [55] demonstrated that kynurenate and methionine levels were upregulated in PanIN but were decreased in PC at the tissue level. LaConti and colleagues identified at least 50 differences in serum metabolites among early PanINs, late PanINs and invasive PC lesions. The data showed that a panel of metabolites achieved a diagnostic accuracy of 81.5% and 73.2% when distinguishing controls from late PanIN and PC cases, respectively [56].
Although circulating metabolites have shown promising diagnostic value in a series of studies, the application of serum metabolomics for the detection of PC remains to be tested and verified in a larger population including patients with various types of pancreatic disease [57]. However, current approaches for analyzing circulating metabolites are not standardized; thus, there may be high heterogeneity and bias among different studies. Furthermore, intricate connections and regulatory nodes among various metabolic processes form a comprehensive metabolic network. Since the concentration of a single metabolite is determined by a number of enzymes and intermediates and usually exhibit a nonlinear quantitative relationship, a panel of metabolic biomarkers might be necessary to establish an effective and stable diagnostic modality.
Autoantibodies
Although immune system dysfunction is common in cancer patients, minor immune responses to a developing tumor can be traced, but such responses are not strong enough to produce clinical manifestations [58,59]. Over the past few years, circulating autoantibodies in the serum of cancer patients have been considered as potential biomarkers for the early detection of cancer. Though little is currently known about the origins and mechanisms of these autoantibodies, data have established that autoantibodies against tumor-associated antigens, such as mutated tumor proteins, misfolded proteins, overexpressed proteins, aberrantly modified proteins and ectopically expressed proteins, are produced in various types of cancer, including PC [30,60].
Bracci et al. [61] analyzed autoantibodies against carboxy-terminal domain, RNA polymerase II, polypeptide A, small phosphatase 1 (CTDSP1), mitogen-activated protein kinase 9 (MAPK9) and nuclear receptor subfamily 2, group E, member 3 (NR2E3) in PC patients (n=300) and controls (n=300). Significant differences in the expression levels of these autoantibodies were observed between PC subjects and controls. However, their diagnostic value was poor, with AUC values less than 0.70. Anti-mucin 1 (MUC1) antibodies have become one focus of autoantibody research in PC. MUC1 is a membrane-associated glycoprotein that is overexpressed in multiple cancers, including PC, and is associated with CA19-9 antigens. Gold et al. [62] reported that a monoclonal antibody against MUC1 had a sensitivity of 77% and a specificity of 95% for differentiating PC patients from normal controls.
Other autoantibodies have also been discovered in the past 10 years, including autoantibodies to two acidic isoforms of the glycolytic enzyme enolase (ENOA1/2) [63], Ezrin [64], a vimentin isoform [65], and calreticulin isoforms [66]. Assisted by the development of mass spectrometry techniques and high-throughput proteomics analysis, a series of autoantibodies, such as those against metabolic enzymes (phosphoglycerate kinase-1, triosephosphate isomerase and isocitrate dehydrogenase) and cytoskeletal proteins, was recently identified [67,68], but their validity as a diagnostic marker remains to be verified in future studies.
Researchers have observed substantial alterations in malignancies that produce novel and immunogenic protein sequences called neoantigens, which are absent from the normal human genome. These neo-epitopes are primarily created by tumor-specific mutations in the genome and become potential targets for cancer detection and immunotherapy [69]. The discovery of additional novel tumor-related autoantibodies is driven by the concept that autoantibodies can indirectly reflect altered genetics or proteomics. However, the frequency at which selected autoantibodies can be detected in cancer patients is low (usually <30%). Tumor heterogeneity complicates the analysis and application of autoantibodies for diagnostic purposes.
Cell-free DNA (cfDNA) and its epigenetic modifications
Nucleic acids can be released into circulation through cellular apoptosis and necrosis, and the size of such nucleic acids ranges primarily from 70 bp to 200 bp. Compared with the healthy population, patients with solid tumors have significantly elevated levels of cfDNA (180 ng/ml on average) [70]. Although the presence of circulating nucleic acids was discovered in the 1940s [71], it was not until recently that interest in them piqued and rapid advancements in their detection and characterization were made.
With the advent of high-throughput sequencing techniques and droplet digital PCR [72,73], we now possess a deeper understanding of tumorigenesis and malignant progression through the acquisition of mutations and epigenetic modifications in cfDNA. In contrast to normal cfDNA, circulating tumor DNA (ctDNA) usually carries mutations that have been reported to match somatic mutations in the primary tumor [74]. Moreover, ctDNA is believed to reflect tumor heterogeneity and to correlate with tumor burden [75-77]. Analyzing ctDNA samples from liquid biopsies could provide insight on the tumor state and genetics.
Somatic mutation of cfDNA
In PC, the predominant genetic characteristic is the high rate of KRAS mutations. Analysis of the genomic landscape of PC has revealed that inactivating mutations in tumor suppressor genes (e.g., CDKN2A, TP53, SMAD4, and BRCA2) are also common [43]. Most PC patients harbor KRAS mutations that can be detected in serum, pancreatic juice, and feces irrespective of clinical stage, even in patients with premalignant pancreatic lesions [78,79]. KRAS mutations are not specific to PC but are also present in various types of malignancies and in chronic pancreatitis [79].
The role of KRAS mutations in chronic pancreatitis and PanIN before PC development has been explored in the last century. According to a thorough investigation of somatic mutations in PanINs, more than 90% of PanIN cases possess KRAS mutations, and the mutation rate is significantly correlated with PanIN grade [80]. These observations suggest that KRAS mutations are an early event during tumorigenesis. However, whether these mutations can be detected in the circulation and utilized for the diagnosis and follow-up of patients is currently unclear. Mulcahy et al. [81] detected KRAS mutations in the plasma of all four patients with chronic pancreatitis (4/4) and reported that all the patients were diagnosed with PC during follow-up in a prospective study. In contrast, Maire et al. [82] reported that KRAS mutations in cfDNA were detected in only 4 of 31 patients with chronic pancreatitis. None of the four patients with KRAS mutations was diagnosed with PC after a mean follow-up of 36 months. Therefore, KRAS mutations are potential biomarkers with considerable sensitivity for the early diagnosis of PC; however, this finding requires further characterization and verification verified in a large study.
In PC, the presence of mutant KRAS has been shown to be related to clinical stage and metastasis [83]. Uemura et al. [84] reported the detection of KRAS mutations in 35% of plasma samples from PC patients before surgery with a polymerase chain reaction combined with restriction fragment length polymorphism (PCR-RFLP) approach. More recently, Bettegowda et al. analyzed 640 plasma samples from patients with various types and stages of cancer, including 155 PC patients, and showed that the detection rate of ctDNA was 48% in patients with resectable PC. In addition, there was a positive correlation between detection of ctDNA and clinical stage in a subgroup analysis [85]. The author also observed a concordance rate of 95% between plasma and primary tumor mutation status. A similar result was obtained by Sausen et al., who reported a ctDNA detection rate of 43% (specificity >99.9%) in 51 patients with localized PC [86]. Moreover, correlations have been found between KRAS mutations and clinicopathological features (e.g., metastasis and overall survival) [87,88].
Because mutations in TP53 and SMAD4 are relatively rare in PC compared with KRAS mutations, few studies have evaluated their diagnostic value in cfDNA. It has been shown that TP53 and SMAD4 mutations are genetic events that occur during the latter stages of disease and are thus most commonly found in PC and grade 3 PanIN [89].
Epigenetic modifications of cfDNA
In addition to genetic abnormalities, aberrant epigenetic modification, especially alterations in the methylation pattern, is notably common in malignancies. Sufficient studies have discovered global hypomethylation of tumor genomic DNA and hypermethylation of tumor suppressor genes. Due to novel methodologies that allow the analysis of epigenetic modifications in DNA from small amounts of sample [90], detection of epigenetic patterns in cfDNA has emerged as a potential biomarker for the early diagnosis of PC.
Previous studies have discovered several targets (UCHL1, NPTX2, SARP2, CLDN5, FOXE1, CDH3, etc.) of abnormal DNA methylation in PC [91]. Similarly, abnormal methylation profiles can be detected in specific regions of cfDNA. Jiao et al. [92] found that the p16 and preproenkephalin promoters were hypermethylated in plasma DNA, with a detection rate of 30% and 25%, respectively, in PC patients. Park et al. [93] reported that the quantitative analysis of plasma NPTX2 by methylation-specific PCR helped differentiate PC from other benign pancreatic lesions, including chronic pancreatitis, with a sensitivity of 80% and a specificity of 75%.
Researchers have also tried to optimize the efficacy of detecting epigenetic patterns in cfDNA by combining several targets. Liggett et al. [94] determined the methylation status of cfDNA in 30 plasma samples from patients with chronic pancreatitis or PC and healthy controls. After statistical analysis, Liggett et al. proposed a panel of 17 gene promoters for differentiating chronic pancreatitis patients from controls (sensitivity =81.7%, specificity =78%) and from PC patients (sensitivity =91.2%, specificity =90.8%) based on the methylation status. Another investigation reported a sensitivity and specificity of 76% and 59%, respectively, in distinguishing PC patients from healthy subjects using the methylation profile of a gene set containing CCND2, SOCS1, and THBS1 [95]. Yi et al. [96] also reported that the promoter methylation status of BNC1 and ADAMTS1 in cfDNA is a promising biomarker for detecting early-stage PC, with a sensitivity of 81% and a specificity of 85% when evaluated together.
Although the analysis of epigenetic alterations in circulating DNA is not as widely used as the detection of KRAS mutations in the diagnosis of PC, the former technique offers some advantages over genetic and serum biomarkers [97,98]. First, the incidence of aberrant DNA methylation at select CpG islands was found to be higher than the incidence of genetic mutations, thereby producing fewer false-negatives. Second, several studies support the concept that aberrant epigenetic alteration is an early event during tumorigenesis that leads to gain-of-function or loss-of-function of critical molecules in cancer cells. Last, the DNA methylation status is relatively stable and can be easily detected with great sensitivity, even when the sample is contaminated.
Cell-free noncoding RNA (ncRNA)
The use of high-throughput genome-wide sequencing technologies in recent years has revealed that the constitution of the human genome and transcriptome is more complicated than previously considered. The human genome comprises genes that can be translated into proteins, which accounts for only 2% of the whole genome, and a large proportion of sequences (at least 75%) that encode noncoding RNAs (ncRNAs) [99]. ncRNAs form a large family that includes microRNAs (miRNAs), small interfering RNAs (siRNAs), piwi-interacting RNAs (piRNAs), small Cajal body-specific RNAs (sdRNAs), and long noncoding RNAs (lncRNAs) [100]. Accumulating evidence has indicated that these ncRNAs play regulatory roles by modifying gene expression at multiple levels through interactions with DNA, RNA, and protein during physiological processes and tumor development [101].
A number of studies have discovered that ncRNA profiles, especially miRNA and lncRNA profiles, are tumor (and PC) specific [102] and can be easily detected in the circulation of patients as potential diagnostic biomarkers of malignancy.
miRNAs
Among circulating ncRNAs, miRNAs have been the subject of the most studies; these small ncRNAs (22-25 nt) play important roles in various physiological processes by binding to complementary mRNA and inhibiting gene expression. Some studies have found evidence that some miRNAs can function as tumor promoters or suppressors. Alterations in miRNA expression have been observed in numerous diseases and in the development and progression of malignancies [103]. Furthermore, miRNAs can circulate in the blood as free RNA bound to hAgo2 and can be incorporated in microparticles such as exosomes [104], which protects the miRNAs from degradation by RNase.
Abnormal expression of certain miRNAs, such as miR-10, miR-21, miR-22, and miR-155, in PC has been demonstrated in disease states according to miRNA profiling studies [105-107]. After several years of exploration, a number of studies have reported that circulating miRNAs or panels of miRNAs identified in the plasma or serum of PC patients show potential diagnostic value, with superiority over CA19-9 [108-110].
Schultz et al. [110] conducted the largest case-control study to date on circulating miRNA in patients with pancreatic diseases, including 409 individuals with PC and 25 patients with chronic pancreatitis, and 312 healthy controls. All the blood samples were collected before the patients underwent chemotherapy or surgery. The authors found nine miRNAs that were consistently dysregulated in the cohort after testing more than 700 miRNAs and identified a panel of miRNAs with diagnostic value that include miR-145, miR-150, miR-223 and miR-636. This diagnostic panel showed an AUC of 0.93, a sensitivity of 0.85, and a specificity of 0.85 but was not superior to CA19-9 (AUC of 0.90, sensitivity of 0.86, and specificity of 0.99). Another multicenter research study conducted by Xu J and colleagues revealed that miR-486-5p exhibits diagnostic potential in discriminating patients with PC from those with chronic pancreatitis (AUC=0.693), pancreatic neuroendocrine tumors (AUC=0.660), and other pancreatic tumors (AUC=0.618) and from healthy controls (AUC=0.861); these values indicate a comparable diagnostic value to that of CA19-9 [108]. The research group also discovered two panels composed of 6 and 3 miRNAs and showed that both panels could accurately distinguish patients with PC from those with chronic pancreatitis (AUC of 0.891 and 0.889) [111]. Using microRNA arrays, Li et al. [112] evaluated 735 circulating miRNAs in sera from patients with PC and controls and identified several miRNAs with an AUC>0.7 for distinguishing PC patients from healthy controls. Of these candidate miRNAs, miR-1290 had the best diagnostic performance in discriminating patients with PC from healthy controls (AUC=0.96), patients with chronic pancreatitis (AUC=0.81), and patients with pancreatic neuroendocrine tumors (AUC=0.80). Moreover, miR-1290 performed better than CA19-9 in distinguishing patients with early-stage PC from controls and was elevated in patients with IPMNs compared with healthy subjects. The results suggest that miR-1290 is a promising circulating miRNA biomarker for the early detection of PC. Likewise, multiple studies have discovered various candidate miRNAs, such as miR-21, miR-18a, miR-155, miR-192, miR-196, miR-200, miR-210, and miR-375 [113-117].
For the early detection of PC, identifying alterations in miRNA profiles of PanIN and IPMN, early events in pancreatic tumorigenesis, provides insight regarding these processes [118,119]. Yu et al. [120] detected 700 miRNAs in PanIN-3 lesions and found that 35 showed altered expression. More attention should be paid to miR-21, miR-155, miR-196 and miR-210 because recent studies have shown their overexpression in PanIN and IPMN lesions [118,119,121-123], suggesting that they could be promising biomarkers for the detection of precancerous lesions. Although different studies have reported aberrant miRNA expression in tissue during the early stages of PC development, few studies have discussed the association between preneoplastic conditions and circulating miRNAs [124], probably because the progression of PC is imperceptible and asymptomatic, which increase the difficulty in screening and collecting samples for testing. Further studies are warranted to explore the value of miRNAs in screening and following patients with PanIN or IPMN.
Above all, circulating miRNAs have advantages, such as small size, stability, convenient extraction and easy detection, but their limitations cannot be ignored. First, it is difficult to quantify and standardize miRNA levels, as there is currently no accepted internal control, making this method unfit for monitoring disease progression. Second, alterations in miRNA profiles are dynamic and easily influenced, leading to potential interference by other diseases or conditions. The origin and mechanisms of circulating RNAs remain unclear, and a panel of miRNAs is recommended in this context to ensure a credible diagnostic result.
LncRNAs
LncRNAs are RNAs longer than 200 nucleotides that are transcribed from multiple locations of the genome with no or limited protein-coding capability [125]. Notably, recent evidence has suggested that lncRNAs are restricted to specific cell types and play crucial roles during tumorigenesis by modulating key pathways at the transcriptional, post-transcriptional, and epigenetic levels [125,126]. Certain lncRNAs could serve as novel biomarkers for the diagnosis of PC and especially for distinguishing different types and subtypes of malignancies based on tissue specificity [127].
Using a cDNA microarray platform, Tahira et al. were the first to explore lncRNA profiling in PC. The authors found that lncRNAs were differentially expressed in primary and metastatic PC lesions, suggesting that lncRNAs may be an indicator of tumor progression [128]. Moreover, recent studies have reported the dysregulation of several lncRNAs, including H19 [129], HOTAIR [130], HOTTIP [131], MALAT-1 [132], HULC [133], and GAS5 [134], in PC lesions compared with normal pancreatic tissue.
The diagnostic performance of circulating lncRNAs has been demonstrated in prostate cancer [135], hepatocellular carcinoma [136], colorectal cancer [137], and non-small cell lung cancer [138]. However, explorations into the association between circulating lncRNAs and PC remain at the primary stage. Only a few pilot studies have tested the diagnostic value of lncRNAs in sera of patients with PC. Wang et al. [139] reported that plasma HDRF and RDRF, fragments of the lncRNAs HOTTIP-005 and RP11-567G11.1, are potential biomarkers; HDRF alone, RDRF alone, and a combination of HDRF and RDRF have AUC values of 0.867, 0.770, and 0.862, respectively, in distinguishing PC patients from healthy controls. We believe that circulating lncRNAs will assist in the early diagnosis of PC in the near future.
CTCs
Although the existence of CTCs was reported by Ashworth nearly one century ago [140], it was not until recently that the selective enrichment and precise analysis of CTCs have become possible, providing a thorough understanding of the underlying mechanisms and associations between CTCs and malignancies. Several studies have shown that CTCs can enter the bloodstream through the shedding of local tumor cells from the primary lesion in the early stages of tumor development and that CTCs can be detected before metastases are established [141-143], suggesting an opportunity for early detection. Moreover, CTCs carry both tumor markers on the surface and somatic mutations [144], thereby enabling ‘‘real-time biopsies’’ of cancer.
Several studies have identified CTCs in 40%-100% of patients with PC [145]. The clinical value of CTCs in predicting PC metastasis and prognosis was recently shown [146]. However, their roles in the early detection and diagnosis of PC have not been extensively explored.
Technological advances have accompanied the discovery of CTCs. Based on the phenomenon that CTCs usually express epithelial cell adhesion molecule (EpCAM), these cells can be selected from peripheral whole blood samples using the CellSearch platform [147]. A study by de Albuquerque et al. [148] reported the presence of CTCs in 47% of PC patients (n=34) and in no healthy controls (n=40), as evidenced by immunomagnetic enrichment using antibodies against EpCAM followed by RT-PCR. However, in another study using the CellSearch system, one or more CTCs were detected in only 5% of locally advanced non-metastatic PC patients (n=79) [149]. Another technique, isolation by the size of epithelial tumor cells (ISET), is a filtration-based, marker-independent method for CTC capture that has, on average, a higher CTC detection rate than CellSearch (93% vs. 40%) [150]. Using a filtration-based method and KRAS mutational analysis, Kulemann et al. [151] detected CTCs in 73% of 11 patients with PC regardless of tumor stage. The detection rate was 75% (3 of 4) among patients with early PC (American Joint Committee on Cancer stage I-II), and no CTCs were found in blood samples from 9 healthy controls. Recent progress in detection techniques has yielded a higher CTC detection rate. Yang et al. [152] applied a newly developed platform, integrated subtraction enrichment and immunostaining-fluorescence in situ hybridization, to analyze pancreatic CTCs, with a reported sensitivity of 88% and a specificity of 90% at a cutoff value of 2 cells/7.5 ml for the diagnosis of PC. For early PC (AJCC stage I-II), the CTC detection rate was 12/13. In another study, blood samples from PC patients were analyzed using the microfluidic NanoVelcro CTC chip. The investigator reported the presence of CTCs in 54/72 patients with confirmed PC, resulting in a sensitivity of 75.0%, a specificity of 96.4%, and an AUC of 0.867 [153].
In addition to CTC morphology and immunologic surface markers, some studies have focused on substrates, such as mRNA and DNA mutations, inside CTCs. Previous studies have detected CEA mRNA in CTCs with RT-PCR and reported the sensitivity (33.3%-75.0%) and specificity (94.6%-96%) for the detection of PC [154-156]. Epithelial-associated molecule CK-20 mRNA was also shown to be a potential biomarker for the detection of PC-associated CTCs [157]. Ankeny et al. [153] analyzed KRAS mutations in CTCs and primary tumor tissue from five patients with PC and found 100% concordance for KRAS mutation subtype between the primary tumor tissue and CTCs.
Though CTCs show promise in the early detection of PC, the performance of this diagnostic approach relies heavily on detection methodology. Therefore, the establishment of a standardized detection method and large-scale validation are urgently required before clinical application.
Exosomes
Exosomes, which are small vesicles released from the plasma membrane by almost all cells, including cancer cells, have been shown to play an important role in intercellular communication, tumorigenesis and cancer metastasis [158-160]. Structurally, exosomes are enveloped by a lipid bilayer membrane with tissue-specific content instead of cellular organelles, such as pathogenic mRNA, miRNA, DNA fragments, and proteins. After release, exosomes are stable in the extracellular environment or circulation and can be taken up by neighboring or distant cells [161]. During this transfer process, exosomes exchange material and information between cells, inducing gene expression or mediating RNA silencing [162]. Recent studies have implicated PC-derived exosomes in the early development of PC [161] and showed that they are beneficial for establishing a premetastatic niche in the liver [163] and that they promote tumor formation and proliferation [164]. Considering their vital functions and extensive distribution, exosomes are potential circulating biomarkers for the detection of PC.
Exosomes contain a variety of proteins, nucleic acids, and lipids from cancer cells that are candidate diagnostic biomarkers. Several proteins were reported to be highly expressed in PC-derived exosomes [165]. Of these proteins, Melo and colleagues [166] chose glypican-1 (GPC1), a membrane-anchored protein, for further study because of its overexpression in both PC and IPMN and its roles in tumor promotion. This exosomal marker exhibited an essentially perfect performance with an AUC of 1.0 in distinguishing PC patients from healthy controls and from patients with benign pancreatic diseases. GPC1-positive exosomes were also detected at the precancerous lesion stage, such as at the PanIN and IPMN stages. Interestingly, GPC1 from serum exosomes showed superior sensitivity and specificity to freely circulating GPC1 from whole serum. Thus, the diagnostic performance of GPC1 is probably attributed to its enrichment in exosomes, thereby highlighting the potential of exosomes and their contents as biomarkers for the early detection of PC.
Studies thus far have provided sufficient evidence for the diagnostic and prognostic significance of exosomal miRNA in malignancies [167]. miRNAs in circulating PC-derived exosomes have been shown to be differentially expressed compared with non-malignant control exosomes and have been implicated in several biological properties of cancer [168-171]. A study conducted by Madhavan and colleagues [172] examined the diagnostic value of a panel of five proteins (CD44v6, Tspan8, EpCAM, MET, and CD104) and four miRNAs (miR-1246, miR-4644, miR-3976, and miR-4306) in circulating exosomes. The combination of these biomarkers could discriminate PC from non-PC cases, including healthy controls, chronic pancreatitis patients, and benign pancreatic neoplasm patients, with a sensitivity of 1.00 and a specificity of 0.80.
Circulating exosomes from PC patients contain a large proportion of tumor DNA, which harbors DNA copy number variations, high-frequency somatic mutations and expressed fusion genes [173]. A recent study by Yang et al. [174] explored the potential clinical utility of circulating exosomal DNA for the identification of KRAS and TP53 mutations. The detection rate of the KRASG12D mutation in exosomal DNA was 39.6%, 28.6%, and 2.6% in patients with PC, patients with IPMN, and healthy controls, respectively; however, TP53R273H mutations were detected in 4.2% of the PC cases and none of the healthy subjects. Another study involving 263 individuals evaluated exosomal KRAS mutations in all stages of PC and compared the detection rate in exosomes with that in cfDNA [175]. This previous study reported that more KRAS mutations were identified in exosomal DNA than in cfDNA from PC patients and that fewer such mutations were identified in exosomal DNA than in cfDNA from age-matched controls. Moreover, mutant KRAS in exosomal DNA was detected in 43.6% of early-stage PC patients, suggesting its promising utility in the early diagnosis of PC.
In general, exosomes have some unique advantages. Exosomes are widely distributed in nearly all body fluids, including serum, and are relatively stable when stored long term at -80°C. Second, PC-derived exosomes enter the circulation at an early stage of cancer development and are related to metastasis. These findings are critical for the early detection of PC because PC cells are able to metastasize at an early stage, which greatly influences prognosis. Last, exosomes contain a combination of proteins, DNA, coding and noncoding RNAs, and lipids that can be used as a natural panel of biomarkers for simultaneous evaluation. In summary, exosomes are promising biomarkers for the early detection of PC, although more evidence from large-scale validation studies are required prior to clinical application.
Conclusion and perspectives
An ideal diagnostic method for PC should definitively distinguish malignant lesions from benign lesions, provide precise tumor staging, and detect early-stage disease and preneoplastic conditions. Though it takes years or decades for PanIN lesions to progress to PC, thus providing a time frame for diagnosis and an opportunity for early management, there are many challenges in the early detection of PC, including its asymptomatic nature, the lack of a characteristic radiological manifestation, and the absence of specific molecules in body fluid. Therefore, convenient and highly sensitive diagnostic tests for screening PC are probably more important than tests with good specificity but moderate sensitivity. Primary screening using circulating biomarkers followed by confirmative diagnosis based on imaging and pathologic results might be the future strategy for diagnosing PC.
Due to technical advances in recent years, a variety of novel biomarkers from body fluids, including blood, urine, saliva, pancreatic juice, and stool, has been discovered in studies on the early detection of PC [176,177]. Circulating biomarkers from blood have advantages in terms of stability, convenience, and abundance compared with traditional imaging workups and have exhibited a better diagnostic performance than biomarkers from other body fluids. Moreover, a series of biomarkers can be detected and analyzed efficiently, providing a comprehensive overview of PC.
Various circulating biomarkers have been widely studied, but some limitations are present in most studies. These issues should be handled with caution in future research. First, tumor heterogeneity has been recognized to complicate accurate diagnoses. One or two biomarkers can hardly provide a comprehensive diagnosis of cancer in the context of precision medicine. Additionally, suboptimal sample selection may lead to misinterpretation of the diagnostic value. Most samples in many studies were collected from patients with advanced disease rather than from those with early disease. Adequate controls, including samples from patients with chronic pancreatitis, autoimmune pancreatitis, pancreatic neuroendocrine tumors, or cystadenoma, should also be collected to evaluate the validity of the differential diagnosis. Third, dynamic changes in biomarkers need to be monitored during follow-up or after treatment, especially in high-risk populations. Last, researchers should not ignore the impact of comorbidities. Obstructive jaundice and diabetes are frequently seen in patients with PC, and these conditions have been shown to influence the performance of biomarkers [178,179].
Considering these situations, one option is to develop a diagnostic panel based on a series of biomarkers. Although CA19-9 has been reported to have worse diagnostic performance than several circulating biomarkers in small-scale studies, it remains the only circulating biomarker that is widely accepted and used for the diagnosis of PC. New biomarkers discovered in future studies should be compared to and combined with CA19-9 in a panel to improve sensitivity and specificity. The lack of sufficient samples from noninvasive precursor lesions and early-stage PC must be addressed, and animal models are important tools for research. Genetically engineered mouse models harboring key mutations such as those in KRAS and TP53 spontaneously develop PanIN and PC [180,181]. These models can be used to identify biomarkers in PanIN and early-stage PC, to better understand tumorigenesis and other malignant behaviors, and to test novel therapeutics. In-depth exploration of the fundamentals of PC development and the nature of precursor lesions is definitely a critical step toward discovering and applying novel biomarkers. Collaborative large-scale prospective studies are required in the future to ascertain the reliability of potential biomarkers.
There is still a long journey from bench to bedside. Hopefully, circulating biomarkers will greatly benefit PC patients in the near future by providing the opportunity for early diagnosis and a better prognosis.
Acknowledgements
This study was supported by the National Natural Science Foundation of China (Grant Number 81772555).
Disclosure of conflict of interest
None.
References
- 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68:7–30. doi: 10.3322/caac.21442. [DOI] [PubMed] [Google Scholar]
- 2.Ghaneh P, Costello E, Neoptolemos JP. Biology and management of pancreatic cancer. Gut. 2007;56:1134–1152. doi: 10.1136/gut.2006.103333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Costello E, Greenhalf W, Neoptolemos JP. New biomarkers and targets in pancreatic cancer and their application to treatment. Nat Rev Gastroenterol Hepatol. 2012;9:435–444. doi: 10.1038/nrgastro.2012.119. [DOI] [PubMed] [Google Scholar]
- 4.Neoptolemos JP, Stocken DD, Friess H, Bassi C, Dunn JA, Hickey H, Beger H, Fernandez-Cruz L, Dervenis C, Lacaine F, Falconi M, Pederzoli P, Pap A, Spooner D, Kerr DJ, Buchler MW European Study Group for Pancreatic Cancer. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med. 2004;350:1200–1210. doi: 10.1056/NEJMoa032295. [DOI] [PubMed] [Google Scholar]
- 5.Neoptolemos JP, Stocken DD, Bassi C, Ghaneh P, Cunningham D, Goldstein D, Padbury R, Moore MJ, Gallinger S, Mariette C, Wente MN, Izbicki JR, Friess H, Lerch MM, Dervenis C, Olah A, Butturini G, Doi R, Lind PA, Smith D, Valle JW, Palmer DH, Buckels JA, Thompson J, McKay CJ, Rawcliffe CL, Buchler MW European Study Group for Pancreatic Cancer. Adjuvant chemotherapy with fluorouracil plus folinic acid vs. gemcitabine following pancreatic cancer resection: a randomized controlled trial. JAMA. 2010;304:1073–1081. doi: 10.1001/jama.2010.1275. [DOI] [PubMed] [Google Scholar]
- 6.Vincent A, Herman J, Schulick R, Hruban RH, Goggins M. Pancreatic cancer. Lancet. 2011;378:607–620. doi: 10.1016/S0140-6736(10)62307-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, Kamiyama M, Hruban RH, Eshleman JR, Nowak MA, Velculescu VE, Kinzler KW, Vogelstein B, Iacobuzio-Donahue CA. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467:1114–1117. doi: 10.1038/nature09515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Rulyak SJ, Lowenfels AB, Maisonneuve P, Brentnall TA. Risk factors for the development of pancreatic cancer in familial pancreatic cancer kindreds. Gastroenterology. 2003;124:1292–1299. doi: 10.1016/s0016-5085(03)00272-5. [DOI] [PubMed] [Google Scholar]
- 9.Bunger S, Laubert T, Roblick UJ, Habermann JK. Serum biomarkers for improved diagnostic of pancreatic cancer: a current overview. J Cancer Res Clin Oncol. 2011;137:375–389. doi: 10.1007/s00432-010-0965-x. [DOI] [PubMed] [Google Scholar]
- 10.Mahadevan D, Von Hoff DD. Tumor-stroma interactions in pancreatic ductal adenocarcinoma. Mol Cancer Ther. 2007;6:1186–1197. doi: 10.1158/1535-7163.MCT-06-0686. [DOI] [PubMed] [Google Scholar]
- 11.Steinberg WM, Barkin JS, Bradley EL 3rd, DiMagno E, Layer P, Canto MI, Levy MJ. Should patients with a strong family history of pancreatic cancer be screened on a periodic basis for cancer of the pancreas? Pancreas. 2009;38:e137–150. doi: 10.1097/MPA.0b013e3181a86b2c. [DOI] [PubMed] [Google Scholar]
- 12.Pan S, Chen R, Crispin DA, May D, Stevens T, McIntosh MW, Bronner MP, Ziogas A, Anton-Culver H, Brentnall TA. Protein alterations associated with pancreatic cancer and chronic pancreatitis found in human plasma using global quantitative proteomics profiling. J Proteome Res. 2011;10:2359–2376. doi: 10.1021/pr101148r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Locker GY, Hamilton S, Harris J, Jessup JM, Kemeny N, Macdonald JS, Somerfield MR, Hayes DF, Bast RC Jr ASCO. ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J. Clin. Oncol. 2006;24:5313–5327. doi: 10.1200/JCO.2006.08.2644. [DOI] [PubMed] [Google Scholar]
- 14.Wong D, Ko AH, Hwang J, Venook AP, Bergsland EK, Tempero MA. Serum CA19-9 decline compared to radiographic response as a surrogate for clinical outcomes in patients with metastatic pancreatic cancer receiving chemotherapy. Pancreas. 2008;37:269–274. doi: 10.1097/MPA.0b013e31816d8185. [DOI] [PubMed] [Google Scholar]
- 15.Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74:2913–2921. doi: 10.1158/0008-5472.CAN-14-0155. [DOI] [PubMed] [Google Scholar]
- 16.Singh S, Tang SJ, Sreenarasimhaiah J, Lara LF, Siddiqui A. The clinical utility and limitations of serum carbohydrate antigen (CA19-9) as a diagnostic tool for pancreatic cancer and cholangiocarcinoma. Dig Dis Sci. 2011;56:2491–2496. doi: 10.1007/s10620-011-1709-8. [DOI] [PubMed] [Google Scholar]
- 17.Gold DV, Gaedcke J, Ghadimi BM, Goggins M, Hruban RH, Liu M, Newsome G, Goldenberg DM. PAM4 enzyme immunoassay alone and in combination with CA 19-9 for the detection of pancreatic adenocarcinoma. Cancer. 2013;119:522–528. doi: 10.1002/cncr.27762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Huang Z, Liu F. Diagnostic value of serum carbohydrate antigen 19-9 in pancreatic cancer: a meta-analysis. Tumour Biol. 2014;35:7459–7465. doi: 10.1007/s13277-014-1995-9. [DOI] [PubMed] [Google Scholar]
- 19.Rosty C, Goggins M. Early detection of pancreatic carcinoma. Hematol Oncol Clin North Am. 2002;16:37–52. doi: 10.1016/s0889-8588(01)00007-7. [DOI] [PubMed] [Google Scholar]
- 20.Goggins M. Molecular markers of early pancreatic cancer. J. Clin. Oncol. 2005;23:4524–4531. doi: 10.1200/JCO.2005.19.711. [DOI] [PubMed] [Google Scholar]
- 21.Brand RE, Nolen BM, Zeh HJ, Allen PJ, Eloubeidi MA, Goldberg M, Elton E, Arnoletti JP, Christein JD, Vickers SM, Langmead CJ, Landsittel DP, Whitcomb DC, Grizzle WE, Lokshin AE. Serum biomarker panels for the detection of pancreatic cancer. Clin Cancer Res. 2011;17:805–816. doi: 10.1158/1078-0432.CCR-10-0248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Faca VM, Song KS, Wang H, Zhang Q, Krasnoselsky AL, Newcomb LF, Plentz RR, Gurumurthy S, Redston MS, Pitteri SJ, Pereira-Faca SR, Ireton RC, Katayama H, Glukhova V, Phanstiel D, Brenner DE, Anderson MA, Misek D, Scholler N, Urban ND, Barnett MJ, Edelstein C, Goodman GE, Thornquist MD, McIntosh MW, DePinho RA, Bardeesy N, Hanash SM. A mouse to human search for plasma proteome changes associated with pancreatic tumor development. PLoS Med. 2008;5:e123. doi: 10.1371/journal.pmed.0050123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Chang ST, Zahn JM, Horecka J, Kunz PL, Ford JM, Fisher GA, Le QT, Chang DT, Ji H, Koong AC. Identification of a biomarker panel using a multiplex proximity ligation assay improves accuracy of pancreatic cancer diagnosis. J Transl Med. 2009;7:105. doi: 10.1186/1479-5876-7-105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Makawita S, Dimitromanolakis A, Soosaipillai A, Soleas I, Chan A, Gallinger S, Haun RS, Blasutig IM, Diamandis EP. Validation of four candidate pancreatic cancer serological biomarkers that improve the performance of CA19.9. BMC Cancer. 2013;13:404. doi: 10.1186/1471-2407-13-404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Park HD, Kang ES, Kim JW, Lee KT, Lee KH, Park YS, Park JO, Lee J, Heo JS, Choi SH, Choi DW, Kim S, Lee JK, Lee SY. Serum CA19-9, cathepsin D, and matrix metalloproteinase-7 as a diagnostic panel for pancreatic ductal adenocarcinoma. Proteomics. 2012;12:3590–3597. doi: 10.1002/pmic.201200101. [DOI] [PubMed] [Google Scholar]
- 26.Zhang Y, Yang J, Li H, Wu Y, Zhang H, Chen W. Tumor markers CA19-9, CA242 and CEA in the diagnosis of pancreatic cancer: a meta-analysis. Int J Clin Exp Med. 2015;8:11683–11691. [PMC free article] [PubMed] [Google Scholar]
- 27.Luo G, Liu C, Guo M, Cheng H, Lu Y, Jin K, Liu L, Long J, Xu J, Lu R, Ni Q, Yu X. Potential biomarkers in lewis negative patients with pancreatic cancer. Ann Surg. 2017;265:800–805. doi: 10.1097/SLA.0000000000001741. [DOI] [PubMed] [Google Scholar]
- 28.Yue T, Maupin KA, Fallon B, Li L, Partyka K, Anderson MA, Brenner DE, Kaul K, Zeh H, Moser AJ, Simeone DM, Feng Z, Brand RE, Haab BB. Enhanced discrimination of malignant from benign pancreatic disease by measuring the CA 19-9 antigen on specific protein carriers. PLoS One. 2011;6:e29180. doi: 10.1371/journal.pone.0029180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Partyka K, Maupin KA, Brand RE, Haab BB. Diverse monoclonal antibodies against the CA 19-9 antigen show variation in binding specificity with consequences for clinical interpretation. Proteomics. 2012;12:2212–2220. doi: 10.1002/pmic.201100676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kaur S, Baine MJ, Jain M, Sasson AR, Batra SK. Early diagnosis of pancreatic cancer: challenges and new developments. Biomark Med. 2012;6:597–612. doi: 10.2217/bmm.12.69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Pistol-Tanase C, Raducan E, Dima SO, Albulescu L, Alina I, Marius P, Cruceru LM, Codorean E, Neagu TM, Popescu I. Assessment of soluble angiogenic markers in pancreatic cancer. Biomark Med. 2008;2:447–455. doi: 10.2217/17520363.2.5.447. [DOI] [PubMed] [Google Scholar]
- 32.Zeh HJ, Winikoff S, Landsittel DP, Gorelik E, Marrangoni AM, Velikokhatnaya L, Winans MT, Lee K, Moser A, Bartlett D, Lotze MT, Siegfried JM, Whitcomb D, Papacristou G, Slivka A, Bigbee WL, Lokshin AE. Multianalyte profiling of serum cytokines for detection of pancreatic cancer. Cancer Biomark. 2005;1:259–269. doi: 10.3233/cbm-2005-1601. [DOI] [PubMed] [Google Scholar]
- 33.Shaw VE, Lane B, Jenkinson C, Cox T, Greenhalf W, Halloran CM, Tang J, Sutton R, Neoptolemos JP, Costello E. Serum cytokine biomarker panels for discriminating pancreatic cancer from benign pancreatic disease. Mol Cancer. 2014;13:114. doi: 10.1186/1476-4598-13-114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wingren C, Sandstrom A, Segersvard R, Carlsson A, Andersson R, Lohr M, Borrebaeck CA. Identification of serum biomarker signatures associated with pancreatic cancer. Cancer Res. 2012;72:2481–2490. doi: 10.1158/0008-5472.CAN-11-2883. [DOI] [PubMed] [Google Scholar]
- 35.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 36.Commisso C, Davidson SM, Soydaner-Azeloglu RG, Parker SJ, Kamphorst JJ, Hackett S, Grabocka E, Nofal M, Drebin JA, Thompson CB, Rabinowitz JD, Metallo CM, Vander Heiden MG, Bar-Sagi D. Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature. 2013;497:633–637. doi: 10.1038/nature12138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, Perera RM, Ferrone CR, Mullarky E, Shyh-Chang N, Kang Y, Fleming JB, Bardeesy N, Asara JM, Haigis MC, DePinho RA, Cantley LC, Kimmelman AC. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature. 2013;496:101–105. doi: 10.1038/nature12040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Perera RM, Stoykova S, Nicolay BN, Ross KN, Fitamant J, Boukhali M, Lengrand J, Deshpande V, Selig MK, Ferrone CR, Settleman J, Stephanopoulos G, Dyson NJ, Zoncu R, Ramaswamy S, Haas W, Bardeesy N. Transcriptional control of autophagy-lysosome function drives pancreatic cancer metabolism. Nature. 2015;524:361–365. doi: 10.1038/nature14587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Garber K. Energy boost: the Warburg effect returns in a new theory of cancer. J Natl Cancer Inst. 2004;96:1805–1806. doi: 10.1093/jnci/96.24.1805. [DOI] [PubMed] [Google Scholar]
- 40.Spratlin JL, Serkova NJ, Eckhardt SG. Clinical applications of metabolomics in oncology: a review. Clin Cancer Res. 2009;15:431–440. doi: 10.1158/1078-0432.CCR-08-1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Denkert C, Bucher E, Hilvo M, Salek R, Oresic M, Griffin J, Brockmoller S, Klauschen F, Loibl S, Barupal DK, Budczies J, Iljin K, Nekljudova V, Fiehn O. Metabolomics of human breast cancer: new approaches for tumor typing and biomarker discovery. Genome Med. 2012;4:37. doi: 10.1186/gm336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Vermeersch KA, Styczynski MP. Applications of metabolomics in cancer research. J Carcinog. 2013;12:9. doi: 10.4103/1477-3163.113622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, Miller DK, Wilson PJ, Patch AM, Wu J, Chang DK, Cowley MJ, Gardiner BB, Song S, Harliwong I, Idrisoglu S, Nourse C, Nourbakhsh E, Manning S, Wani S, Gongora M, Pajic M, Scarlett CJ, Gill AJ, Pinho AV, Rooman I, Anderson M, Holmes O, Leonard C, Taylor D, Wood S, Xu Q, Nones K, Fink JL, Christ A, Bruxner T, Cloonan N, Kolle G, Newell F, Pinese M, Mead RS, Humphris JL, Kaplan W, Jones MD, Colvin EK, Nagrial AM, Humphrey ES, Chou A, Chin VT, Chantrill LA, Mawson A, Samra JS, Kench JG, Lovell JA, Daly RJ, Merrett ND, Toon C, Epari K, Nguyen NQ, Barbour A, Zeps N Australian Pancreatic Cancer Genome Initiative. Kakkar N, Zhao F, Wu YQ, Wang M, Muzny DM, Fisher WE, Brunicardi FC, Hodges SE, Reid JG, Drummond J, Chang K, Han Y, Lewis LR, Dinh H, Buhay CJ, Beck T, Timms L, Sam M, Begley K, Brown A, Pai D, Panchal A, Buchner N, De Borja R, Denroche RE, Yung CK, Serra S, Onetto N, Mukhopadhyay D, Tsao MS, Shaw PA, Petersen GM, Gallinger S, Hruban RH, Maitra A, Iacobuzio-Donahue CA, Schulick RD, Wolfgang CL, Morgan RA, Lawlor RT, Capelli P, Corbo V, Scardoni M, Tortora G, Tempero MA, Mann KM, Jenkins NA, Perez-Mancera PA, Adams DJ, Largaespada DA, Wessels LF, Rust AG, Stein LD, Tuveson DA, Copeland NG, Musgrove EA, Scarpa A, Eshleman JR, Hudson TJ, Sutherland RL, Wheeler DA, Pearson JV, McPherson JD, Gibbs RA, Grimmond SM. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012;491:399–405. doi: 10.1038/nature11547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ozdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, Laklai H, Sugimoto H, Kahlert C, Novitskiy SV, De Jesus-Acosta A, Sharma P, Heidari P, Mahmood U, Chin L, Moses HL, Weaver VM, Maitra A, Allison JP, LeBleu VS, Kalluri R. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25:719–734. doi: 10.1016/j.ccr.2014.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, Dekleva EN, Saunders T, Becerra CP, Tattersall IW, Westphalen CB, Kitajewski J, Fernandez-Barrena MG, Fernandez-Zapico ME, Iacobuzio-Donahue C, Olive KP, Stanger BZ. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25:735–747. doi: 10.1016/j.ccr.2014.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Sousa CM, Biancur DE, Wang X, Halbrook CJ, Sherman MH, Zhang L, Kremer D, Hwang RF, Witkiewicz AK, Ying H, Asara JM, Evans RM, Cantley LC, Lyssiotis CA, Kimmelman AC. Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion. Nature. 2016;536:479–483. doi: 10.1038/nature19084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zhou W, Capello M, Fredolini C, Racanicchi L, Piemonti L, Liotta LA, Novelli F, Petricoin EF. Proteomic analysis reveals Warburg effect and anomalous metabolism of glutamine in pancreatic cancer cells. J Proteome Res. 2012;11:554–563. doi: 10.1021/pr2009274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kobayashi T, Nishiumi S, Ikeda A, Yoshie T, Sakai A, Matsubara A, Izumi Y, Tsumura H, Tsuda M, Nishisaki H, Hayashi N, Kawano S, Fujiwara Y, Minami H, Takenawa T, Azuma T, Yoshida M. A novel serum metabolomics-based diagnostic approach to pancreatic cancer. Cancer Epidemiol Biomarkers Prev. 2013;22:571–579. doi: 10.1158/1055-9965.EPI-12-1033. [DOI] [PubMed] [Google Scholar]
- 49.Ritchie SA, Akita H, Takemasa I, Eguchi H, Pastural E, Nagano H, Monden M, Doki Y, Mori M, Jin W, Sajobi TT, Jayasinghe D, Chitou B, Yamazaki Y, White T, Goodenowe DB. Metabolic system alterations in pancreatic cancer patient serum: potential for early detection. BMC Cancer. 2013;13:416. doi: 10.1186/1471-2407-13-416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zhang G, He P, Tan H, Budhu A, Gaedcke J, Ghadimi BM, Ried T, Yfantis HG, Lee DH, Maitra A, Hanna N, Alexander HR, Hussain SP. Integration of metabolomics and transcriptomics revealed a fatty acid network exerting growth inhibitory effects in human pancreatic cancer. Clin Cancer Res. 2013;19:4983–4993. doi: 10.1158/1078-0432.CCR-13-0209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Di Gangi IM, Mazza T, Fontana A, Copetti M, Fusilli C, Ippolito A, Mattivi F, Latiano A, Andriulli A, Vrhovsek U, Pazienza V. Metabolomic profile in pancreatic cancer patients: a consensus-based approach to identify highly discriminating metabolites. Oncotarget. 2016;7:5815–5829. doi: 10.18632/oncotarget.6808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Sakai A, Suzuki M, Kobayashi T, Nishiumi S, Yamanaka K, Hirata Y, Nakagawa T, Azuma T, Yoshida M. Pancreatic cancer screening using a multiplatform human serum metabolomics system. Biomark Med. 2016;10:577–586. doi: 10.2217/bmm-2016-0020. [DOI] [PubMed] [Google Scholar]
- 53.Leichtle AB, Ceglarek U, Weinert P, Nakas CT, Nuoffer JM, Kase J, Conrad T, Witzigmann H, Thiery J, Fiedler GM. Pancreatic carcinoma, pancreatitis, and healthy controls: metabolite models in a three-class diagnostic dilemma. Metabolomics. 2013;9:677–687. doi: 10.1007/s11306-012-0476-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Yuan C, Clish CB, Wu C, Mayers JR, Kraft P, Townsend MK, Zhang M, Tworoger SS, Bao Y, Qian ZR, Rubinson DA, Ng K, Giovannucci EL, Ogino S, Stampfer MJ, Gaziano JM, Ma J, Sesso HD, Anderson GL, Cochrane BB, Manson JE, Torrence ME, Kimmelman AC, Amundadottir LT, Vander Heiden MG, Fuchs CS, Wolpin BM. Circulating metabolites and survival among patients with pancreatic cancer. J Natl Cancer Inst. 2016;108:djv409. doi: 10.1093/jnci/djv409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Wen S, Li Z, Feng J, Bai J, Lin X, Huang H. Metabonomic changes from pancreatic intraepithelial neoplasia to pancreatic ductal adenocarcinoma in tissues from rats. Cancer Sci. 2016;107:836–845. doi: 10.1111/cas.12939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.LaConti JJ, Laiakis EC, Mays AD, Peran I, Kim SE, Shay JW, Riegel AT, Fornace AJ Jr, Wellstein A. Distinct serum metabolomics profiles associated with malignant progression in the KrasG12D mouse model of pancreatic ductal adenocarcinoma. BMC Genomics. 2015;16(Suppl 1):S1. doi: 10.1186/1471-2164-16-S1-S1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Shakour S, Ruffin MT, Zick SM, Lubman DM, Nie S. Serum metabolomic analysis of pancreatic cancer--letter. Cancer Epidemiol Biomarkers Prev. 2013;22:1921. doi: 10.1158/1055-9965.EPI-13-0672. [DOI] [PubMed] [Google Scholar]
- 58.Baldwin RW. Tumour-specific immunity against spontaneous rat tumours. Int J Cancer. 1966;1:257–264. doi: 10.1002/ijc.2910010305. [DOI] [PubMed] [Google Scholar]
- 59.Kobold S, Lutkens T, Cao Y, Bokemeyer C, Atanackovic D. Autoantibodies against tumor-related antigens: incidence and biologic significance. Hum Immunol. 2010;71:643–651. doi: 10.1016/j.humimm.2010.03.015. [DOI] [PubMed] [Google Scholar]
- 60.Desmetz C, Mange A, Maudelonde T, Solassol J. Autoantibody signatures: progress and perspectives for early cancer detection. J Cell Mol Med. 2011;15:2013–2024. doi: 10.1111/j.1582-4934.2011.01355.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Bracci PM, Zhou M, Young S, Wiemels J. Serum autoantibodies to pancreatic cancer antigens as biomarkers of pancreatic cancer in a San Francisco Bay Area case-control study. Cancer. 2012;118:5384–5394. doi: 10.1002/cncr.27538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Gold DV, Modrak DE, Ying Z, Cardillo TM, Sharkey RM, Goldenberg DM. New MUC1 serum immunoassay differentiates pancreatic cancer from pancreatitis. J. Clin. Oncol. 2006;24:252–258. doi: 10.1200/JCO.2005.02.8282. [DOI] [PubMed] [Google Scholar]
- 63.Tomaino B, Cappello P, Capello M, Fredolini C, Sperduti I, Migliorini P, Salacone P, Novarino A, Giacobino A, Ciuffreda L, Alessio M, Nistico P, Scarpa A, Pederzoli P, Zhou W, Petricoin Iii EF, Liotta LA, Giovarelli M, Milella M, Novelli F. Circulating autoantibodies to phosphorylated alpha-enolase are a hallmark of pancreatic cancer. J Proteome Res. 2011;10:105–112. doi: 10.1021/pr100213b. [DOI] [PubMed] [Google Scholar]
- 64.Capello M, Cappello P, Linty FC, Chiarle R, Sperduti I, Novarino A, Salacone P, Mandili G, Naccarati A, Sacerdote C, Beghelli S, Bersani S, Barbi S, Bassi C, Scarpa A, Nistico P, Giovarelli M, Vineis P, Milella M, Novelli F. Autoantibodies to Ezrin are an early sign of pancreatic cancer in humans and in genetically engineered mouse models. J Hematol Oncol. 2013;6:67. doi: 10.1186/1756-8722-6-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Hong SH, Misek DE, Wang H, Puravs E, Hinderer R, Giordano TJ, Greenson JK, Brenner DE, Simeone DM, Logsdon CD, Hanash SM. Identification of a specific vimentin isoform that induces an antibody response in pancreatic cancer. Biomark Insights. 2006;1:175–183. [PMC free article] [PubMed] [Google Scholar]
- 66.Hong SH, Misek DE, Wang H, Puravs E, Giordano TJ, Greenson JK, Brenner DE, Simeone DM, Logsdon CD, Hanash SM. An autoantibody-mediated immune response to calreticulin isoforms in pancreatic cancer. Cancer Res. 2004;64:5504–5510. doi: 10.1158/0008-5472.CAN-04-0077. [DOI] [PubMed] [Google Scholar]
- 67.Patwa TH, Li C, Poisson LM, Kim HY, Pal M, Ghosh D, Simeone DM, Lubman DM. The identification of phosphoglycerate kinase-1 and histone H4 autoantibodies in pancreatic cancer patient serum using a natural protein microarray. Electrophoresis. 2009;30:2215–2226. doi: 10.1002/elps.200800857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Tomaino B, Cappello P, Capello M, Fredolini C, Ponzetto A, Novarino A, Ciuffreda L, Bertetto O, De Angelis C, Gaia E, Salacone P, Milella M, Nistico P, Alessio M, Chiarle R, Giuffrida MG, Giovarelli M, Novelli F. Autoantibody signature in human ductal pancreatic adenocarcinoma. J Proteome Res. 2007;6:4025–4031. doi: 10.1021/pr070281a. [DOI] [PubMed] [Google Scholar]
- 69.Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74. doi: 10.1126/science.aaa4971. [DOI] [PubMed] [Google Scholar]
- 70.Cui Q, Lewis IA, Hegeman AD, Anderson ME, Li J, Schulte CF, Westler WM, Eghbalnia HR, Sussman MR, Markley JL. Metabolite identification via the Madison Metabolomics Consortium Database. Nat Biotechnol. 2008;26:162–164. doi: 10.1038/nbt0208-162. [DOI] [PubMed] [Google Scholar]
- 71.Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, Knippers R. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 2001;61:1659–1665. [PubMed] [Google Scholar]
- 72.Earl J, Garcia-Nieto S, Martinez-Avila JC, Montans J, Sanjuanbenito A, Rodrguez-Garrote M, Lisa E, Menda E, Lobo E, Malats N, Carrato A, Guillen-Ponce C. 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. doi: 10.1186/s12885-015-1779-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Zill OA, Greene C, Sebisanovic D, Siew LM, Leng J, Vu M, Hendifar AE, Wang Z, Atreya CE, Kelley RK, Van Loon K, Ko AH, Tempero MA, Bivona TG, Munster PN, Talasaz A, Collisson EA. Cell-free DNA next-generation sequencing in pancreatobiliary carcinomas. Cancer Discov. 2015;5:1040–1048. doi: 10.1158/2159-8290.CD-15-0274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.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. doi: 10.1002/cncr.29364. [DOI] [PubMed] [Google Scholar]
- 75.Diaz LA Jr, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J. Clin. Oncol. 2014;32:579–586. doi: 10.1200/JCO.2012.45.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Fleischhacker M, Schmidt B. Circulating nucleic acids (CNAs) and cancer--a survey. Biochim Biophys Acta. 2007;1775:181–232. doi: 10.1016/j.bbcan.2006.10.001. [DOI] [PubMed] [Google Scholar]
- 77.Newman AM, Bratman SV, To J, Wynne JF, Eclov NC, Modlin LA, Liu CL, Neal JW, Wakelee HA, Merritt RE, Shrager JB, Loo BW Jr, Alizadeh AA, Diehn M. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med. 2014;20:548–554. doi: 10.1038/nm.3519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Kamiyama H, Jimeno A, Hong SM, Fu B, Lin MT, Calhoun ES, Kamiyama M, Walter K, Nikolskaya T, Nikolsky Y, Hartigan J, Smith DR, Hidalgo M, Leach SD, Klein AP, Jaffee EM, Goggins M, Maitra A, Iacobuzio-Donahue C, Eshleman JR, Kern SE, Hruban RH, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–1806. doi: 10.1126/science.1164368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Yanagisawa A, Ohtake K, Ohashi K, Hori M, Kitagawa T, Sugano H, Kato Y. Frequent c-Ki-ras oncogene activation in mucous cell hyperplasias of pancreas suffering from chronic inflammation. Cancer Res. 1993;53:953–956. [PubMed] [Google Scholar]
- 80.Kanda M, Matthaei H, Wu J, Hong SM, Yu J, Borges M, Hruban RH, Maitra A, Kinzler K, Vogelstein B, Goggins M. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology. 2012;142:730–733. e739. doi: 10.1053/j.gastro.2011.12.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Mulcahy HE, Lyautey J, Lederrey C, qi Chen X, Anker P, Alstead EM, Ballinger A, Farthing MJ, Stroun M. A prospective study of K-ras mutations in the plasma of pancreatic cancer patients. Clin Cancer Res. 1998;4:271–275. [PubMed] [Google Scholar]
- 82.Maire F, Micard S, Hammel P, Voitot H, Levy 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. doi: 10.1038/sj.bjc.6600475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Castells A, Puig P, Mora J, Boadas J, Boix L, Urgell E, Sole M, Capella G, Lluis F, Fernandez-Cruz L, Navarro S, Farre A. 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. doi: 10.1200/JCO.1999.17.2.578. [DOI] [PubMed] [Google Scholar]
- 84.Uemura T, Hibi K, Kaneko T, Takeda S, Inoue S, Okochi O, Nagasaka T, Nakao A. Detection of K-ras mutations in the plasma DNA of pancreatic cancer patients. J Gastroenterol. 2004;39:56–60. doi: 10.1007/s00535-003-1245-1. [DOI] [PubMed] [Google Scholar]
- 85.Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, Bartlett BR, Wang H, Luber B, Alani RM, Antonarakis ES, Azad NS, Bardelli A, Brem H, Cameron JL, Lee CC, Fecher LA, Gallia GL, Gibbs P, Le D, Giuntoli RL, Goggins M, Hogarty MD, Holdhoff M, Hong SM, Jiao Y, Juhl HH, Kim JJ, Siravegna G, Laheru DA, Lauricella C, Lim M, Lipson EJ, Marie SK, Netto GJ, Oliner KS, Olivi A, Olsson L, Riggins GJ, Sartore-Bianchi A, Schmidt K, Shih lM, Oba-Shinjo SM, Siena S, Theodorescu D, Tie J, Harkins TT, Veronese S, Wang TL, Weingart JD, Wolfgang CL, Wood LD, Xing D, Hruban RH, Wu J, Allen PJ, Schmidt CM, Choti MA, Velculescu VE, Kinzler KW, Vogelstein B, Papadopoulos N, Diaz LA Jr. Detection of circulating tumor dna in early-and late-stage human malignancies. Sci Transl Med. 2014;6:224ra224. doi: 10.1126/scitranslmed.3007094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Sausen M, Phallen J, Adleff V, Jones S, Leary RJ, Barrett MT, Anagnostou V, Parpart-Li S, Murphy D, Kay Li Q, Hruban CA, Scharpf R, White JR, O’Dwyer PJ, Allen PJ, Eshleman JR, Thompson CB, Klimstra DS, Linehan DC, Maitra A, Hruban RH, Diaz LA Jr, Von Hoff DD, Johansen JS, Drebin JA, Velculescu VE. Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients. Nat Commun. 2015;6:7686. doi: 10.1038/ncomms8686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.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. doi: 10.1002/cncr.29364. [DOI] [PubMed] [Google Scholar]
- 88.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. doi: 10.3109/07357907.2014.1001894. [DOI] [PubMed] [Google Scholar]
- 89.Maitra A, Adsay NV, Argani P, Iacobuzio-Donahue C, De Marzo A, Cameron JL, Yeo CJ, Hruban RH. Multicomponent analysis of the pancreatic adenocarcinoma progression model using a pancreatic intraepithelial neoplasia tissue microarray. Mod Pathol. 2003;16:902–912. doi: 10.1097/01.MP.0000086072.56290.FB. [DOI] [PubMed] [Google Scholar]
- 90.Levenson VV, Melnikov AA. The MethDet: a technology for biomarker development. Expert Rev Mol Diagn. 2011;11:807–812. doi: 10.1586/erm.11.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Sato N, Fukushima N, Maitra A, Matsubayashi H, Yeo CJ, Cameron JL, Hruban RH, Goggins M. Discovery of novel targets for aberrant methylation in pancreatic carcinoma using high-throughput microarrays. Cancer Res. 2003;63:3735–3742. [PubMed] [Google Scholar]
- 92.Jiao L, Zhu J, Hassan MM, Evans DB, Abbruzzese JL, Li D. K-ras mutation and p16 and preproenkephalin promoter hypermethylation in plasma DNA of pancreatic cancer patients: in relation to cigarette smoking. Pancreas. 2007;34:55–62. doi: 10.1097/01.mpa.0000246665.68869.d4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Park JK, Ryu JK, Yoon WJ, Lee SH, Lee GY, Jeong KS, Kim YT, Yoon YB. The role of quantitative NPTX2 hypermethylation as a novel serum diagnostic marker in pancreatic cancer. Pancreas. 2012;41:95–101. doi: 10.1097/MPA.0b013e318221c903. [DOI] [PubMed] [Google Scholar]
- 94.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. doi: 10.1002/cncr.24893. [DOI] [PubMed] [Google Scholar]
- 95.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. doi: 10.1002/jso.21208. [DOI] [PubMed] [Google Scholar]
- 96.Yi JM, Guzzetta AA, Bailey VJ, Downing SR, Van Neste L, Chiappinelli KB, Keeley BP, Stark A, Herrera A, Wolfgang C, Pappou EP, Iacobuzio-Donahue CA, Goggins MG, Herman JG, Wang TH, Baylin SB, Ahuja N. Novel methylation biomarker panel for the early detection of pancreatic cancer. Clin Cancer Res. 2013;19:6544–6555. doi: 10.1158/1078-0432.CCR-12-3224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Laird PW. The power and the promise of DNA methylation markers. Nat Rev Cancer. 2003;3:253–266. doi: 10.1038/nrc1045. [DOI] [PubMed] [Google Scholar]
- 98.Miyamoto K, Ushijima T. Diagnostic and therapeutic applications of epigenetics. Jpn J Clin Oncol. 2005;35:293–301. doi: 10.1093/jjco/hyi088. [DOI] [PubMed] [Google Scholar]
- 99.Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, Xue C, Marinov GK, Khatun J, Williams BA, Zaleski C, Rozowsky J, Roder M, Kokocinski F, Abdelhamid RF, Alioto T, Antoshechkin I, Baer MT, Bar NS, Batut P, Bell K, Bell I, Chakrabortty S, Chen X, Chrast J, Curado J, Derrien T, Drenkow J, Dumais E, Dumais J, Duttagupta R, Falconnet E, Fastuca M, Fejes-Toth K, Ferreira P, Foissac S, Fullwood MJ, Gao H, Gonzalez D, Gordon A, Gunawardena H, Howald C, Jha S, Johnson R, Kapranov P, King B, Kingswood C, Luo OJ, Park E, Persaud K, Preall JB, Ribeca P, Risk B, Robyr D, Sammeth M, Schaffer L, See LH, Shahab A, Skancke J, Suzuki AM, Takahashi H, Tilgner H, Trout D, Walters N, Wang H, Wrobel J, Yu Y, Ruan X, Hayashizaki Y, Harrow J, Gerstein M, Hubbard T, Reymond A, Antonarakis SE, Hannon G, Giddings MC, Ruan Y, Wold B, Carninci P, Guigo R, Gingeras TR. Landscape of transcription in human cells. Nature. 2012;489:101–108. doi: 10.1038/nature11233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.St Laurent G, Wahlestedt C, Kapranov P. The Landscape of long noncoding RNA classification. Trends Genet. 2015;31:239–251. doi: 10.1016/j.tig.2015.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Prensner JR, Chinnaiyan AM. The emergence of lncRNAs in cancer biology. Cancer Discov. 2011;1:391–407. doi: 10.1158/2159-8290.CD-11-0209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Muller S, Raulefs S, Bruns P, Afonso-Grunz F, Plotner A, Thermann R, Jager C, Schlitter AM, Kong B, Regel I, Roth WK, Rotter B, Hoffmeier K, Kahl G, Koch I, Theis FJ, Kleeff J, Winter P, Michalski CW. Next-generation sequencing reveals novel differentially regulated mRNAs, lncRNAs, miRNAs, sdRNAs and a piRNA in pancreatic cancer. Mol Cancer. 2015;14:94. doi: 10.1186/s12943-015-0358-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, Downing JR, Jacks T, Horvitz HR, Golub TR. MicroRNA expression profiles classify human cancers. Nature. 2005;435:834–838. doi: 10.1038/nature03702. [DOI] [PubMed] [Google Scholar]
- 104.Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–659. doi: 10.1038/ncb1596. [DOI] [PubMed] [Google Scholar]
- 105.Lee EJ, Gusev Y, Jiang J, Nuovo GJ, Lerner MR, Frankel WL, Morgan DL, Postier RG, Brackett DJ, Schmittgen TD. Expression profiling identifies microRNA signature in pancreatic cancer. Int J Cancer. 2007;120:1046–1054. doi: 10.1002/ijc.22394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Bloomston M, Frankel WL, Petrocca F, Volinia S, Alder H, Hagan JP, Liu CG, Bhatt D, Taccioli C, Croce CM. MicroRNA expression patterns to differentiate pancreatic adenocarcinoma from normal pancreas and chronic pancreatitis. JAMA. 2007;297:1901–1908. doi: 10.1001/jama.297.17.1901. [DOI] [PubMed] [Google Scholar]
- 107.Roldo C, Missiaglia E, Hagan JP, Falconi M, Capelli P, Bersani S, Calin GA, Volinia S, Liu CG, Scarpa A, Croce CM. MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. J. Clin. Oncol. 2006;24:4677–4684. doi: 10.1200/JCO.2005.05.5194. [DOI] [PubMed] [Google Scholar]
- 108.Xu J, Cao Z, Liu W, You L, Zhou L, Wang C, Lou W, Sun B, Miao Y, Liu X, Zhang T, Zhao Y. Plasma miRNAs effectively distinguish patients with pancreatic cancer from controls: a Multicenter Study. Ann Surg. 2016;263:1173–1179. doi: 10.1097/SLA.0000000000001345. [DOI] [PubMed] [Google Scholar]
- 109.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. doi: 10.1002/ijc.26422. [DOI] [PubMed] [Google Scholar]
- 110.Schultz NA, Dehlendorff C, Jensen BV, Bjerregaard JK, Nielsen KR, Bojesen SE, Calatayud D, Nielsen SE, Yilmaz M, Hollander NH, Andersen KK, Johansen JS. MicroRNA biomarkers in whole blood for detection of pancreatic cancer. JAMA. 2014;311:392–404. doi: 10.1001/jama.2013.284664. [DOI] [PubMed] [Google Scholar]
- 111.Cao Z, Liu C, Xu J, You L, Wang C, Lou W, Sun B, Miao Y, Liu X, Wang X, Zhang T, Zhao Y. Plasma microRNA panels to diagnose pancreatic cancer: results from a multicenter study. Oncotarget. 2016;7:41575–41583. doi: 10.18632/oncotarget.9491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.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. doi: 10.1158/1078-0432.CCR-12-3092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.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. doi: 10.1097/MPA.0b013e318296bb34. [DOI] [PubMed] [Google Scholar]
- 114.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. doi: 10.1593/tlo.09256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.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. doi: 10.1158/0008-5472.CAN-09-4227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Morimura R, Komatsu S, Ichikawa D, Takeshita H, Tsujiura M, Nagata H, Konishi H, Shiozaki A, Ikoma H, Okamoto K, Ochiai T, Taniguchi H, Otsuji E. Novel diagnostic value of circulating miR-18a in plasma of patients with pancreatic cancer. Br J Cancer. 2011;105:1733–1740. doi: 10.1038/bjc.2011.453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Liu M, Du Y, Gao J, Liu J, Kong X, Gong Y, Li Z, Wu H, Chen H. Aberrant expression miR-196a is associated with abnormal apoptosis, invasion, and proliferation of pancreatic cancer cells. Pancreas. 2013;42:1169–1181. doi: 10.1097/MPA.0b013e3182962acb. [DOI] [PubMed] [Google Scholar]
- 118.Xue Y, Abou Tayoun AN, Abo KM, Pipas JM, Gordon SR, Gardner TB, Barth RJ Jr, Suriawinata AA, Tsongalis GJ. MicroRNAs as diagnostic markers for pancreatic ductal adenocarcinoma and its precursor, pancreatic intraepithelial neoplasm. Cancer Genet. 2013;206:217–221. doi: 10.1016/j.cancergen.2013.05.020. [DOI] [PubMed] [Google Scholar]
- 119.du Rieu MC, Torrisani J, Selves J, Al Saati T, Souque A, Dufresne M, Tsongalis GJ, Suriawinata AA, Carrere N, Buscail L, Cordelier P. MicroRNA-21 is induced early in pancreatic ductal adenocarcinoma precursor lesions. Clin Chem. 2010;56:603–612. doi: 10.1373/clinchem.2009.137364. [DOI] [PubMed] [Google Scholar]
- 120.Yu J, Li A, Hong SM, Hruban RH, Goggins M. MicroRNA alterations of pancreatic intraepithelial neoplasias. Clin Cancer Res. 2012;18:981–992. doi: 10.1158/1078-0432.CCR-11-2347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Ryu JK, Hong SM, Karikari CA, Hruban RH, Goggins MG, Maitra A. Aberrant MicroRNA-155 expression is an early event in the multistep progression of pancreatic adenocarcinoma. Pancreatology. 2010;10:66–73. doi: 10.1159/000231984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Caponi S, Funel N, Frampton AE, Mosca F, Santarpia L, Van der Velde AG, Jiao LR, De Lio N, Falcone A, Kazemier G, Meijer GA, Verheul HM, Vasile E, Peters GJ, Boggi U, Giovannetti E. The good, the bad and the ugly: a tale of miR-101, miR-21 and miR-155 in pancreatic intraductal papillary mucinous neoplasms. Ann Oncol. 2013;24:734–741. doi: 10.1093/annonc/mds513. [DOI] [PubMed] [Google Scholar]
- 123.Slater EP, Strauch K, Rospleszcz S, Ramaswamy A, Esposito I, Kloppel G, Matthai E, Heeger K, Fendrich V, Langer P, Bartsch DK. MicroRNA-196a and -196b as potential biomarkers for the early detection of familial pancreatic cancer. Transl Oncol. 2014;7:464–471. doi: 10.1016/j.tranon.2014.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Komatsu S, Ichikawa D, Miyamae M, Kawaguchi T, Morimura R, Hirajima S, Okajima W, Ohashi T, Imamura T, Konishi H, Shiozaki A, Ikoma H, Okamoto K, Taniguchi H, Otsuji E. Malignant potential in pancreatic neoplasm; new insights provided by circulating miR-223 in plasma. Expert Opin Biol Ther. 2015;15:773–785. doi: 10.1517/14712598.2015.1029914. [DOI] [PubMed] [Google Scholar]
- 125.Ulitsky I, Bartel DP. lincRNAs: genomics, evolution, and mechanisms. Cell. 2013;154:26–46. doi: 10.1016/j.cell.2013.06.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Quinn JJ, Chang HY. Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet. 2016;17:47–62. doi: 10.1038/nrg.2015.10. [DOI] [PubMed] [Google Scholar]
- 127.Flippot R, Malouf GG, Su X, Mouawad R, Spano JP, Khayat D. Cancer subtypes classification using long non-coding RNA. Oncotarget. 2016;7:54082–54093. doi: 10.18632/oncotarget.10213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.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. doi: 10.1186/1476-4598-10-141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Scaiewicz V, Sorin V, Fellig Y, Birman T, Mizrahi A, Galula J, Abu-Lail R, Shneider T, Ohana P, Buscail L, Hochberg A, Czerniak A. Use of H19 gene regulatory sequences in DNA-based therapy for pancreatic cancer. J Oncol. 2010;2010:178174. doi: 10.1155/2010/178174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.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. doi: 10.1038/onc.2012.193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Fu Z, Chen C, Zhou Q, Wang Y, Zhao Y, Zhao X, Li W, Zheng S, Ye H, Wang L, He Z, Lin Q, Li Z, Chen R. LncRNA HOTTIP modulates cancer stem cell properties in human pancreatic cancer by regulating HOXA9. Cancer Lett. 2017;410:68–81. doi: 10.1016/j.canlet.2017.09.019. [DOI] [PubMed] [Google Scholar]
- 132.Jiao F, Hu H, Yuan C, Wang L, Jiang W, Jin Z, Guo Z, Wang L. Elevated expression level of long noncoding RNA MALAT-1 facilitates cell growth, migration and invasion in pancreatic cancer. Oncol Rep. 2014;32:2485–2492. doi: 10.3892/or.2014.3518. [DOI] [PubMed] [Google Scholar]
- 133.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. doi: 10.1007/s12032-014-0346-4. [DOI] [PubMed] [Google Scholar]
- 134.Lu X, Fang Y, Wang Z, Xie J, Zhan Q, Deng X, Chen H, Jin J, Peng C, Li H, Shen B. Downregulation of gas5 increases pancreatic cancer cell proliferation by regulating CDK6. Cell Tissue Res. 2013;354:891–896. doi: 10.1007/s00441-013-1711-x. [DOI] [PubMed] [Google Scholar]
- 135.Ren S, Wang F, Shen J, Sun Y, Xu W, Lu J, Wei M, Xu C, Wu C, Zhang Z, Gao X, Liu Z, Hou J, Huang J, Sun Y. Long non-coding RNA metastasis associated in lung adenocarcinoma transcript 1 derived miniRNA as a novel plasma-based biomarker for diagnosing prostate cancer. Eur J Cancer. 2013;49:2949–2959. doi: 10.1016/j.ejca.2013.04.026. [DOI] [PubMed] [Google Scholar]
- 136.Yuan W, Sun Y, Liu L, Zhou B, Wang S, Gu D. Circulating LncRNAs serve as diagnostic markers for hepatocellular carcinoma. Cell Physiol Biochem. 2017;44:125–132. doi: 10.1159/000484589. [DOI] [PubMed] [Google Scholar]
- 137.Zhao W, Song M, Zhang J, Kuerban M, Wang H. Combined identification of long non-coding RNA CCAT1 and HOTAIR in serum as an effective screening for colorectal carcinoma. Int J Clin Exp Pathol. 2015;8:14131–14140. [PMC free article] [PubMed] [Google Scholar]
- 138.Tantai J, Hu D, Yang Y, Geng J. Combined identification of long non-coding RNA XIST and HIF1A-AS1 in serum as an effective screening for non-small cell lung cancer. Int J Clin Exp Pathol. 2015;8:7887–7895. [PMC free article] [PubMed] [Google Scholar]
- 139.Wang Y, Li Z, Zheng S, Zhou Y, Zhao L, Ye H, Zhao X, Gao W, Fu Z, Zhou Q, Liu Y, Chen R. Expression profile of long non-coding RNAs in pancreatic cancer and their clinical significance as biomarkers. Oncotarget. 2015;6:35684–35698. doi: 10.18632/oncotarget.5533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Riva F, Dronov OI, Khomenko DI, Huguet F, Louvet C, Mariani P, Stern MH, Lantz O, Proudhon C, Pierga JY, Bidard FC. Clinical applications of circulating tumor DNA and circulating tumor cells in pancreatic cancer. Mol Oncol. 2016;10:481–493. doi: 10.1016/j.molonc.2016.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Husemann Y, Geigl JB, Schubert F, Musiani P, Meyer M, Burghart E, Forni G, Eils R, Fehm T, Riethmuller G, Klein CA. Systemic spread is an early step in breast cancer. Cancer Cell. 2008;13:58–68. doi: 10.1016/j.ccr.2007.12.003. [DOI] [PubMed] [Google Scholar]
- 142.Pantel K, Brakenhoff RH. Dissecting the metastatic cascade. Nat Rev Cancer. 2004;4:448–456. doi: 10.1038/nrc1370. [DOI] [PubMed] [Google Scholar]
- 143.Rhim AD, Mirek ET, Aiello NM, Maitra A, Bailey JM, McAllister F, Reichert M, Beatty GL, Rustgi AK, Vonderheide RH, Leach SD, Stanger BZ. EMT and dissemination precede pancreatic tumor formation. Cell. 2012;148:349–361. doi: 10.1016/j.cell.2011.11.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.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. doi: 10.1016/j.lungcan.2011.10.018. [DOI] [PubMed] [Google Scholar]
- 145.Iwanicki-Caron I, Basile P, Toure E, Antonietti M, Lecleire S, Di Fiore A, Oden-Gangloff A, Blanchard F, Lemoine F, Di Fiore F, Sabourin JC, Michel P. Usefulness of circulating tumor cell detection in pancreatic adenocarcinoma diagnosis. Am J Gastroenterol. 2013;108:152–155. doi: 10.1038/ajg.2012.367. [DOI] [PubMed] [Google Scholar]
- 146.Riva F, Dronov OI, Khomenko DI, Huguet F, Louvet C, Mariani P, Stern MH, Lantz O, Proudhon C, Pierga JY, Bidard FC. Clinical applications of circulating tumor DNA and circulating tumor cells in pancreatic cancer. Mol Oncol. 2016;10:481–493. doi: 10.1016/j.molonc.2016.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D, Ulkus L, Smith MR, Kwak EL, Digumarthy S, Muzikansky A, Ryan P, Balis UJ, Tompkins RG, Haber DA, Toner M. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature. 2007;450:1235–1239. doi: 10.1038/nature06385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.de Albuquerque A, Kubisch I, Breier G, Stamminger G, Fersis N, Eichler A, Kaul S, Stolzel U. Multimarker gene analysis of circulating tumor cells in pancreatic cancer patients: a feasibility study. Oncology. 2012;82:3–10. doi: 10.1159/000335479. [DOI] [PubMed] [Google Scholar]
- 149.Bidard FC, Huguet F, Louvet C, Mineur L, Bouche O, Chibaudel B, Artru P, Desseigne F, Bachet JB, Mathiot C, Pierga JY, Hammel P. Circulating tumor cells in locally advanced pancreatic adenocarcinoma: the ancillary CirCe 07 study to the LAP 07 trial. Ann Oncol. 2013;24:2057–2061. doi: 10.1093/annonc/mdt176. [DOI] [PubMed] [Google Scholar]
- 150.Khoja L, Backen A, Sloane R, Menasce L, Ryder D, Krebs M, Board R, Clack G, Hughes A, Blackhall F, Valle JW, Dive C. A pilot study to explore circulating tumour cells in pancreatic cancer as a novel biomarker. Br J Cancer. 2012;106:508–516. doi: 10.1038/bjc.2011.545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Kulemann B, Pitman MB, Liss AS, Valsangkar N, Fernandez-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. doi: 10.1097/MPA.0000000000000324. [DOI] [PubMed] [Google Scholar]
- 152.Gao Y, Zhu Y, Zhang Z, Zhang C, Huang X, Yuan Z. Clinical significance of pancreatic circulating tumor cells using combined negative enrichment and immunostaining-fluorescence in situ hybridization. J Exp Clin Cancer Res. 2016;35:66. doi: 10.1186/s13046-016-0340-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Ankeny JS, Court CM, Hou S, Li Q, Song M, Wu D, Chen JF, Lee T, Lin M, Sho S, Rochefort MM, Girgis MD, Yao J, Wainberg ZA, Muthusamy VR, Watson RR, Donahue TR, Hines OJ, Reber HA, Graeber TG, Tseng HR, Tomlinson JS. Circulating tumour cells as a biomarker for diagnosis and staging in pancreatic cancer. Br J Cancer. 2016;114:1367–1375. doi: 10.1038/bjc.2016.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.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. doi: 10.1007/BF02573871. [DOI] [PubMed] [Google Scholar]
- 155.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. doi: 10.1158/1078-0432.CCR-03-0130. [DOI] [PubMed] [Google Scholar]
- 156.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. doi: 10.1016/s0002-9610(99)00086-0. [DOI] [PubMed] [Google Scholar]
- 157.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] [Google Scholar]
- 158.Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev Immunol. 2014;14:195–208. doi: 10.1038/nri3622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Melo SA, Sugimoto H, O’Connell JT, Kato N, Villanueva A, Vidal A, Qiu L, Vitkin E, Perelman LT, Melo CA, Lucci A, Ivan C, Calin GA, Kalluri R. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell. 2014;26:707–721. doi: 10.1016/j.ccell.2014.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, Singh S, Williams C, Soplop N, Uryu K, Pharmer L, King T, Bojmar L, Davies AE, Ararso Y, Zhang T, Zhang H, Hernandez J, Weiss JM, Dumont-Cole VD, Kramer K, Wexler LH, Narendran A, Schwartz GK, Healey JH, Sandstrom P, Labori KJ, Kure EH, Grandgenett PM, Hollingsworth MA, de Sousa M, Kaur S, Jain M, Mallya K, Batra SK, Jarnagin WR, Brady MS, Fodstad O, Muller V, Pantel K, Minn AJ, Bissell MJ, Garcia BA, Kang Y, Rajasekhar VK, Ghajar CM, Matei I, Peinado H, Bromberg J, Lyden D. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527:329–335. doi: 10.1038/nature15756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Azmi AS, Bao B, Sarkar FH. Exosomes in cancer development, metastasis, and drug resistance: a comprehensive review. Cancer Metastasis Rev. 2013;32:623–642. doi: 10.1007/s10555-013-9441-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Lo Cicero A, Stahl PD, Raposo G. Extracellular vesicles shuffling intercellular messages: for good or for bad. Curr Opin Cell Biol. 2015;35:69–77. doi: 10.1016/j.ceb.2015.04.013. [DOI] [PubMed] [Google Scholar]
- 163.Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, Becker A, Hoshino A, Mark MT, Molina H, Xiang J, Zhang T, Theilen TM, Garcia-Santos G, Williams C, Ararso Y, Huang Y, Rodrigues G, Shen TL, Labori KJ, Lothe IM, Kure EH, Hernandez J, Doussot A, Ebbesen SH, Grandgenett PM, Hollingsworth MA, Jain M, Mallya K, Batra SK, Jarnagin WR, Schwartz RE, Matei I, Peinado H, Stanger BZ, Bromberg J, Lyden D. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. 2015;17:816–826. doi: 10.1038/ncb3169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Charrier A, Chen R, Chen L, Kemper S, Hattori T, Takigawa M, Brigstock DR. Connective tissue growth factor (CCN2) and microRNA-21 are components of a positive feedback loop in pancreatic stellate cells (PSC) during chronic pancreatitis and are exported in PSC-derived exosomes. J Cell Commun Signal. 2014;8:147–156. doi: 10.1007/s12079-014-0220-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Wang H, Rana S, Giese N, Buchler MW, Zoller M. Tspan8, CD44v6 and alpha6beta4 are biomarkers of migrating pancreatic cancer-initiating cells. Int J Cancer. 2013;133:416–426. doi: 10.1002/ijc.28044. [DOI] [PubMed] [Google Scholar]
- 166.Melo SA, Luecke LB, Kahlert C, Fernandez AF, Gammon ST, Kaye J, LeBleu VS, Mittendorf EA, Weitz J, Rahbari N, Reissfelder C, Pilarsky C, Fraga MF, Piwnica-Worms D, Kalluri R. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature. 2015;523:177–182. doi: 10.1038/nature14581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Zoller M. Pancreatic cancer diagnosis by free and exosomal miRNA. World J Gastrointest Pathophysiol. 2013;4:74–90. doi: 10.4291/wjgp.v4.i4.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Lau C, Kim Y, Chia D, Spielmann N, Eibl G, Elashoff D, Wei F, Lin YL, Moro A, Grogan T, Chiang S, Feinstein E, Schafer C, Farrell J, Wong DT. Role of pancreatic cancer-derived exosomes in salivary biomarker development. J Biol Chem. 2013;288:26888–26897. doi: 10.1074/jbc.M113.452458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.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. doi: 10.1186/1477-7819-11-219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Joshi GK, Deitz-McElyea S, Liyanage T, Lawrence K, Mali S, Sardar R, Korc M. Label-free nanoplasmonic-based short noncoding RNA sensing at attomolar concentrations allows for quantitative and highly specific assay of MicroRNA-10b in biological fluids and circulating exosomes. ACS Nano. 2015;9:11075–11089. doi: 10.1021/acsnano.5b04527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Rana S, Yue S, Stadel D, Zoller M. Toward tailored exosomes: the exosomal tetraspanin web contributes to target cell selection. Int J Biochem Cell Biol. 2012;44:1574–1584. doi: 10.1016/j.biocel.2012.06.018. [DOI] [PubMed] [Google Scholar]
- 172.Madhavan B, Yue S, Galli U, Rana S, Gross W, Muller M, Giese NA, Kalthoff H, Becker T, Buchler MW, Zoller M. 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. doi: 10.1002/ijc.29324. [DOI] [PubMed] [Google Scholar]
- 173.San Lucas FA, Allenson K, Bernard V, Castillo J, Kim DU, Ellis K, Ehli EA, Davies GE, Petersen JL, Li D, Wolff R, Katz M, Varadhachary G, Wistuba I, Maitra A, Alvarez H. Minimally invasive genomic and transcriptomic profiling of visceral cancers by next-generation sequencing of circulating exosomes. Ann Oncol. 2016;27:635–641. doi: 10.1093/annonc/mdv604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Yang S, Che SP, Kurywchak P, Tavormina JL, Gansmo LB, Correa de Sampaio P, Tachezy M, Bockhorn M, Gebauer F, Haltom AR, Melo SA, LeBleu VS, Kalluri R. Detection of mutant KRAS and TP53 DNA in circulating exosomes from healthy individuals and patients with pancreatic cancer. Cancer Biol Ther. 2017;18:158–165. doi: 10.1080/15384047.2017.1281499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Allenson K, Castillo J, San Lucas FA, Scelo G, Kim DU, Bernard V, Davis G, Kumar T, Katz M, Overman MJ, Foretova L, Fabianova E, Holcatova I, Janout V, Meric-Bernstam F, Gascoyne P, Wistuba I, Varadhachary G, Brennan P, Hanash S, Li D, Maitra A, Alvarez H. High prevalence of mutant KRAS in circulating exosome-derived DNA from early-stage pancreatic cancer patients. Ann Oncol. 2017;28:741–747. doi: 10.1093/annonc/mdx004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Debernardi S, Massat NJ, Radon TP, Sangaralingam A, Banissi A, Ennis DP, Dowe T, Chelala C, Pereira SP, Kocher HM, Young BD, Bond-Smith G, Hutchins R, Crnogorac-Jurcevic T. Noninvasive urinary miRNA biomarkers for early detection of pancreatic adenocarcinoma. Am J Cancer Res. 2015;5:3455–3466. [PMC free article] [PubMed] [Google Scholar]
- 177.Yang JY, Sun YW, Liu DJ, Zhang JF, Li J, Hua R. MicroRNAs in stool samples as potential screening biomarkers for pancreatic ductal adenocarcinoma cancer. Am J Cancer Res. 2014;4:663–673. [PMC free article] [PubMed] [Google Scholar]
- 178.Tonack S, Jenkinson C, Cox T, Elliott V, Jenkins RE, Kitteringham NR, Greenhalf W, Shaw V, Michalski CW, Friess H, Neoptolemos JP, Costello E. iTRAQ reveals candidate pancreatic cancer serum biomarkers: influence of obstructive jaundice on their performance. Br J Cancer. 2013;108:1846–1853. doi: 10.1038/bjc.2013.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Nie S, Lo A, Wu J, Zhu J, Tan Z, Simeone DM, Anderson MA, Shedden KA, Ruffin MT, Lubman DM. Glycoprotein biomarker panel for pancreatic cancer discovered by quantitative proteomics analysis. J Proteome Res. 2014;13:1873–1884. doi: 10.1021/pr400967x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Mazur PK, Siveke JT. Genetically engineered mouse models of pancreatic cancer: unravelling tumour biology and progressing translational oncology. Gut. 2012;61:1488–1500. doi: 10.1136/gutjnl-2011-300756. [DOI] [PubMed] [Google Scholar]
- 181.Esposito I, Segler A, Steiger K, Kloppel G. Pathology, genetics and precursors of human and experimental pancreatic neoplasms: an update. Pancreatology. 2015;15:598–610. doi: 10.1016/j.pan.2015.08.007. [DOI] [PubMed] [Google Scholar]