Next Article in Journal
Myoferlin Contributes to the Metastatic Phenotype of Pancreatic Cancer Cells by Enhancing Their Migratory Capacity through the Control of Oxidative Phosphorylation
Next Article in Special Issue
Advancing Risk Assessment of Intermediate Risk Prostate Cancer Patients
Previous Article in Journal
Gold Nanorod-Assisted Photothermal Therapy Decreases Bleeding during Breast Cancer Surgery in Dogs and Cats
Previous Article in Special Issue
Determination of PD-L1 Expression in Circulating Tumor Cells of NSCLC Patients and Correlation with Response to PD-1/PD-L1 Inhibitors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Liquid Biopsy Approach for Pancreatic Ductal Adenocarcinoma

by
Etienne Buscail
1,2,
Charlotte Maulat
3,4,
Fabrice Muscari
3,4,
Laurence Chiche
1,2,
Pierre Cordelier
3,
Sandrine Dabernat
1,
Catherine Alix-Panabières
5 and
Louis Buscail
3,6,*
1
INSERM U1035, Bordeaux University, 33000 Bordeaux, France
2
Department of Digestive Surgery, Bordeaux University Hospital, 33600 Pessac, France
3
Université Fédérale Toulouse Midi-Pyrénées, Université Toulouse III Paul Sabatier, INSERM, CRCT, 31330 Toulouse, France
4
Department of Digestive Surgery, Toulouse University Hospital, 31059 Toulouse, France
5
Laboratory of Rare Human Circulating Cells (LCCRH), Montpellier Hospital and University of Montpellier, 34295 Montpellier, France
6
Department of Gastroenterology and Pancreatology, Toulouse University Hospital, 31059 Toulouse, France
*
Author to whom correspondence should be addressed.
Submission received: 8 May 2019 / Revised: 1 June 2019 / Accepted: 14 June 2019 / Published: 19 June 2019
(This article belongs to the Special Issue Liquid Biopsy for Cancer)

Abstract

:
Pancreatic cancer is a public health problem because of its increasing incidence, the absence of early diagnostic tools, and its aggressiveness. Despite recent progress in chemotherapy, the 5-year survival rate remains below 5%. Liquid biopsies are of particular interest from a clinical point of view because they are non-invasive biomarkers released by primary tumours and metastases, remotely reflecting disease burden. Pilot studies have been conducted in pancreatic cancer patients evaluating the detection of circulating tumour cells, cell-free circulating tumour DNA, exosomes, and tumour-educated platelets. There is heterogeneity between the methods used to isolate circulating tumour elements as well as the targets used for their identification. Performances for the diagnosis of pancreatic cancer vary depending of the technique but also the stage of the disease: 30–50% of resectable tumours are positive and 50–100% are positive in locally advanced and/or metastatic cases. A significant prognostic value is demonstrated in 50–70% of clinical studies, irrespective of the type of liquid biopsy. Large prospective studies of homogeneous cohorts of patients are lacking. One way to improve diagnostic and prognostic performances would be to use a combined technological approach for the detection of circulating tumour cells, exosomes, and DNA.

Graphical Abstract

1. Introduction

Pancreatic ductal adenocarcinoma (PDAC) cancer is a significant public health problem and a medical and scientific challenge because of its increasing incidence, the lack of reliable early biomarkers, and the absence of preventive screening and efficient therapies to overcome these aggressive and highly heterogeneous neoplasms [1,2,3]. The only curative treatment is surgery, which is only feasible in 15% of cases. For other patients, the tumour is already metastatic or locally advanced at the time of diagnosis. Despite application of chemotherapy protocols such as FOLFIRINOX (association of 5-fluoro-uracile, oxaliplatin and irinotecan), which increases survival after palliative or adjuvant therapies, the 5-year survival rate remains below 5% [4,5,6]. In parallel with the search for new treatments, several challenges must be faced to help alleviate the dismal prognosis of PDAC. One of these is to discover new biomarkers to ensure early detection, make a meaningful prognosis, and help predict recurrences. Ideally, these questions should all be addressed remotely, without the need for invasive, painful, and risky procedures. In this context, real-time liquid biopsy is an emerging tool of particular interest from the scientific and clinical perspective because complementary circulating biomarkers are released by the tumour and its metastases and therefore distantly reflect the disease [7,8,9,10]. Liquid biopsies are collected in a non-invasive manner and allow diagnosis and molecular follow-up of patients. They have already been implemented in the clinic to monitor and manage the therapies of a number of epithelial cancer patients [11,12].
The purpose of this review is to address all molecular, technological and clinical aspects and issues of liquid biopsies, as they relate to PDAC. Circulating tumour cells (CTCs), cell-free circulating tumour DNA (ctDNA), circulating tumour extracellular vesicles (e.g., exosomes), tumour educated platelets (TEPs), and blood-based protein and metabolite markers are specifically discussed, because they provide pragmatic perspectives on the application of promising technologies to the manage patients at all stages of PDAC.

2. Current Diagnosis for Pancreatic Cancer

PDAC diagnosis usually relies on information obtained using sequential procedures, including imaging data (ultrasonography, computerised tomodensitometry (CT), magnetic resonance imaging (MRI) and endoscopic ultrasound (EUS)). Blood markers are lacking, and the only one used is the carbohydrate antigen 19-9 (CA 19-9) which displays too low sensitivity and specificity to be useful as a stringent diagnostic marker for PDAC. Although CA19-9 is an acceptable marker for advanced and symptomatic tumours (sensitivity of 80%; specificity of 82%), its performance falls rapidly in the case of small non-metastatic lesions [13]. The different imaging techniques aim to detect a pancreatic tissue mass and to determine its spread locally by evaluating possible venous and arterial vascular invasion, but also at distance to detect metastases, specifically peritoneal and hepatic metastases. These are sometimes detected by diffusion MRI. After having assessed its spread, the tumour will be classified as either locally advanced and non-resectable (25% of patients at the time of diagnosis), metastatic (50%), borderline (10%), or resectable (15%). The performance of the CT and the MRI are generally equivalent for the diagnosis and assessment of pancreatic cancer staging [14], with CT more effective for the diagnosis of tumour unresectability. However, in the majority of cases (except for resectable tumours) a pathological confirmation is required either with a fine-needle aspirate of a metastasis or a EUS-guided fine-needle aspiration (EUS-FNA) of the primary tumour. More than 20 years of EUS experience have now paved the way for safe guided FNA biopsies of solid pancreatic lesions for cytopathological analysis [3,15]. EUS-FNA is thus now an effective technique to diagnose and assess the staging of PDAC, especially for tumours less than 2 cm [16,17,18]. However, its performance (and performances of other imaging techniques) greatly depends upon the operators’ experience and certainly on the nature of the PDAC, depending on the importance of stroma. In other words, the more abundant the tumour stroma, the fewer carcinoma cells will be present in EUS-guided microbiopsy samples. Accuracy of EUS-FNA to diagnose malignancy varies widely, with sensitivity ranging from 65% to 95% with a mean accuracy of 85%. In addition, the negative predictive value still ranges from 50% to 70% and the EUS-FNA may be inconclusive or ambiguous in up to 20% of cases [16,19,20] of PDAC. Inconclusive specimens can be defined as the presence of coagulum with normal cells or acellular samples. Ambiguous samples can be defined by the presence of atypia and/or low-grade dysplasia and/or be atypical for the malignancy. Recently, technical and clinical improvements to EUS have been developed such as elastography, contrast-enhanced EUS, and the use of an on-site pathologist. Each of these procedures may improve EUS for tissue characterizations [14,21] as well as for differentiating between PDAC and neuroendocrine tumours, lymphoma, or autoimmune pancreatitis [16,17]. Nevertheless, in cases of strongly suspected PDAC, a negative EUS-FNA result remains problematic. A second EUS-FNA is therefore required, equating to a treatment delay for the patient, which is known to negatively influence the prognosis of PDAC [22]. In addition, there is the problem of the differential diagnosis between PDAC and chronic pancreatitis in its pseudo-tumoural form and, more rarely, with autoimmune pancreatitis. In these cases, histological diagnosis is also crucial to avoid unnecessary surgery.
To assist in cytopathological diagnosis, the research and characterisation of new molecular markers is an active field. Nevertheless, since the oncogenic KRAS point mutation is a frequent event during PDAC, the identification of this mutation in tumour tissues may aid diagnosis. We and others have demonstrated that KRAS-mutation analysis, performed on EUS-FNA samples, appears to be highly accurate in differentiating benign versus malignant pancreatic solid lesions [19,20,23]. Faced with solid pancreatic masses, combining results of the KRAS-mutation assay with cytopathology can increase the sensitivity and accuracy of diagnoses when compared to cytopathology alone [20,24,25,26]. More importantly the negative predictive value of cytopathology alone for this indication is increased from 67% to 88% when combined with a KRAS mutation assay [20,24,25,26]. KRAS mutation detection has also been performed on circulating cell free tumour DNA and on isolated exosomes. The hope is to obtain a non-invasive, reliable, and reproducible biological marker demonstrating the presence of PDAC, irrespective of stage. We will discuss the potential of the different liquid biopsy approaches applicable to PDAC by trying to compare performance, weaknesses, and prospects.

3. Circulating Tumour Cell-Based Diagnosis of Pancreatic Cancer

The clinical utility of CTC detection for cancer patients regained interest during the late 1990s, and is now referred to as liquid biopsy [10]. Progress with regards to CTC capture/identification has predominantly relied on technical advances. Numerous CTC detection methods have been described. These primarily rely on a first step of CTC capture/enrichment based on tumour cell biological (immunological positive or negative selection) or physical properties (size, deformability, cell density), followed by a second detection step based on immunocytochemistry, molecular biology, or a functional assay [27]. Liquid biopsies can be performed on several body fluids, but application to pancreatic cancer detection has mainly been performed on total peripheral blood [28].
At first, liquid biopsies relating to pancreatic cancer used methods combining density centrifugation and RT-PCR detection of tumour markers, which were already known to have a poor specificity as circulating tumour biomarkers. This is exemplified by the low detection rates with CEA mRNA detection (26%, [29]), cytokeratine 20 (CK20, 34%, [30]), and EpCAM (epithelial cell adhesion molecule) (25%, [31]). These low detection rates could not be overcome by recruiting a higher proportion of early stage patients [29,30], except in the case of EpCAM RNA-based detection, since most patients were eligible for up-front surgery (83%, [31]). With increasingly available tools allowing the specific capture of epithelial cells from whole blood, numerous studies have tested combined immunocytochemistry detection of tumour markers on isolated cells. First, the CellSearch© system, currently considered as the gold standard because it has been cleared by the U.S. FDA (Food and drug Administration) for use in metastatic breast, colon, and prostate cancer, has been tested in several studies. This system allowed a PDAC diagnosis to be made in 11–48% of patients in cohorts comprising at least 53% of patients with locally advanced or metastatic disease (Table 1) [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45]. Even when all the patients in the cohort had advanced disease, rates of CTC detection were not observed to increase (with one result for example being 32.3% [39]). As expected, when 100% of the patient cohort comprised resectable cases, the number dropped to less than 7% [42]. These quite disappointing results were usually explained by the fact that the CellSearch® system is based on EpCAM and cytokeratin expression. These epithelial markers are down regulated and might even be lost during the epithelial–mesenchymal transition (EMT, [46,47]), which is a well-accepted fact for CTCs [48]. To limit this detection bias, CTC enrichment of blood samples has been tested using CTC physical properties. The isolation by size of epithelial tumour cells method (ISET, [49]) might be a good alternative for CTC detection, as it is unbiased by the needed presence of cell surface EpCAM. After whole blood cell filtration on a microporous membrane, retained cells are stained by routine histology with or without pan-cytokeratin immunocytochemistry. CTC identification and counting is performed by pathologists trained to identify the specific morphology of tumour cells [49]. In a cohort comprising more than 80% metastatic patients, for which results using both methods were available for 27 patients, the detection rate reached 93% for ISET as compared to only 40% for CellSearch® with higher mean numbers of CTCs (6 vs. 26 CTC/7.5 mL, Table 1). Another cohort including mainly patients with early stage disease found 78% of PDAC [37], even when pan-cytokeratin positivity was associated with CTC detection. The ScreenCell® filtration method identified 67% of PDAC patients, including 72% of advanced diseases, with cytomorphologic criteria after filtration [38,43]. The latter study included the detection of mutant KRAS in CTCs, with high discrepancy between tumour and CTC status. Indeed, whereas 97% of tumours carried mutant KRAS, 18% of the CTCs were found to carry only the KRAS wild type allele. Even CTCs from five out of the 12 metastatic tumours were KRAS WT (Wild Type). Thus, it is possible that cytomorphological-based CTC identification of filtered cells might falsely consider epithelioid or endothelial cells as epithelial tumour cells, leading to a general CTC overestimation. This hypothesis is supported by a report analysing 171 blood samples from patients with various pancreatic diseases (including 63% PDAC, [50]) and nine healthy controls. The nine healthy controls were free of circulating epithelioid cells (CEC), but of the 115 patients with CECs, 25 (15%) had non-malignant diseases. Morphologic characteristics of malignant CECs were undistinguishable from non-malignant CECs. In addition, CECs were also detected in inflammatory benign colonic diseases [51], suggesting that in specific cases only, these methods might detect cells shed by primary tumours as well as by benign lesions. We may also argue that identification of the KRAS mutation, which is detected very early during PDAC oncogenesis, may help detect early disease, which is eligible for surgery, before tumours grow too big or even disseminate.
Most studies nevertheless correlate the presence and/or number of CTCs with clinical parameters (Table 1). As expected, high CTC numbers correlated with metastatic disease [34] with a worse overall or progression free survival [37,38,39,41,42,45]. When sensitivity of detection is low (11%), CTC positivity correlates with adenocarcinoma differentiation [52].
Overall CTC-based diagnostic of PDAC is highly specific, since most studies with healthy control groups report close to 0 false-positive results (except for 3.6% in [35]). However, sensitivity suffers from the rarity of CTCs which are not efficiently captured/enriched by currently available methods. Filtration and morphologic-based CTC identification carries the risk of over interpretation [50]. In summary, the presence of CTCs from resected, locally advanced, or metastatic tumours was associated with poor PDAC patient prognoses, an increased number of metastases and an increased likelihood of relapse. As shown in Table 1, 13 out of 18 studies concluded that the presence of CTCs indicate poor survival. This is also illustrated in Figure 1.

4. Circulating Tumour DNA for Diagnosis and Prognosis of Pancreatic Cancer

ctDNA originates from necrotic or apoptotic cells but can also be actively secreted by cells. CtDNA is highly fragmented with a median size of 170 base pairs, which corresponds to internucleosomal DNA fragments. CtDNA is cell-free, circulating tumour DNA, because it contains mutations that are only specific to cancer cells [9,53]. Only very few studies have investigated methylation, microsatellite instability and allelic imbalance in ctDNA [7,54]. In contrast, the detection of KRAS mutation in plasma and serum appears the most widely used approach [7,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78]. Various effective methods have been developed for KRAS mutation analysis and now replace direct sequencing and other methods requiring a pre-amplification step, such as RFLP. All these new methods include q-PCR methods, allele-specific PCR using amplification refractory mutation system technology or co-amplification at a lower denaturation temperature, PCR methods, pyrosequencing approaches, and real-time PCR methods that use specific probe technologies, such as peptide nucleic acids [62,79,80,81,82]. Digital droplet PCR (dPCR) is another approach which displays exceptional sensitivity and only requires a low starting concentration of DNA template. For example, considering the sensitivity of different KRAS mutation detection techniques (i.e., ratio mutant/wild type KRAS) that of direct sequencing is 10–30%, while that of next generation sequencing (NGS) is 10% and that of dPCR is 0.01% [83,84].
Numerous studies have been conducted to investigate the role of circulating DNA in patients with PDAC. The major studies that included more than 20 patients are compiled in Table 2 [55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,85]. Of the 24 studies, only 13 included patients with benign pancreatic lesions or healthy subjects. Most studies have applied the detection of the KRAS oncogene mutation to identify circulating tumour DNA. Two questions arise with regard to the detection of circulating DNA: Does it have a diagnostic advantage (and/or can it detect early lesions)? Does it have a prognostic value?
It seems that the detection of KRAS mutations from blood still has limited value for the identification of early tumours or micrometastatic disease, which suggests that either a limited amount of ctDNA is released at these stages of the disease, or that the ctDNA concentration is so low and it is so degraded, that its detection requires more sensitive nucleic acid processing and analysis technologies. In the specific context of the PDAC diagnosis, despite the use of the most efficient techniques such as dPCR, there is the problem of sensitivity of the KRAS assay. Indeed, concordance studies have been carried out between the presence of the KRAS mutation in the primary tumour and the search for the mutation in ctDNA: the concordance varies from 25% to 75% and finally the sensitivity of this approach is highly dependent on the nature of the tumour [7,54]. If we analyse the results displayed in Table 2, the presence of a KRAS mutation in ctDNA is observed in nearly 70–80% of locally advanced and metastatic patients while this value ranges from 30% to 68% for patients with resectable tumours. One simple explanation is that the quantity of ctDNA might depend on the number of PDAC cells in the blood as well as the extent of metastasis. However, this postulate remains to be proven. In addition, and as discussed above, the sensitivity is improved in the case of dPCR (43–78%) when compared to simple PCR or sequencing (27–47%) (Table 2).
Several groups, including ours, have investigated whether the presence or absence of a KRAS mutation can influence the prognosis of PDAC [68,86,87,88,89,90]. Biological samples are varied and include tumour tissues, blood, plasma, or EUS-FNA. Overall and regardless of the type of biological sample used, the presence of a KRAS mutation has a negative influence on the prognosis of PDAC patients whether or not they undergo surgery (with complete tumour resection or whether they have locally advanced and/or metastatic PDAC). As shown in Table 2, 17 out of 24 studies concluded that the presence of a KRAS mutation had an adverse effect on survival (Figure 1).
Some studies also pointed out that the KRAS mutational subtype might also negatively influence prognosis per se (such as G12D and G12V) [38,64,68]. A different coupling to the downstream-signalling pathways of the KRAS protein, depending on the type of mutation, may explain these results [91,92,93].
On the whole, it is highly likely that ctDNAs will play a role in PDAC prognosis. However, new prospective studies are required, which specifically include control patient groups (i.e., free of pancreatic disease and not suffering from chronic pancreatitis) in order to clearly evaluate the sensitivity, specificity and predictive values of this assay for a possible non-invasive diagnosis of PDAC. One more issue is the variability of the detection assay itself in the absence of “universal” threshold and quantification values. To gain further insight and reach a definitive conclusion, multicentre studies in a larger (i.e., more than 200 patients), homogeneous cohort of patients (to allow strong multivariate analyses) are certainly needed. These studies must include sequential sampling of blood in order to establish the real reproducibility of the methods and to evaluate the potential performance, to either predict the response to treatment or the likelihood of relapse. The fragmentation partner of ctDNA itself can help distinguish healthy donor from patients with cancer, including pancreatic adenocarcinoma [94]. Indeed, profiles of healthy individuals reflected nucleosomal patterns of white blood cells, whereas patients with cancer have largely altered fragmentation profiles. Using artificial intelligence, researchers were able to detect breast, colorectal, lung, ovarian, pancreatic, gastric or bile duct cancer inpatients ranging from 57% to more than 99% among the seven cancer types at 98% specificity, with an overall area under the curve value of 0.94. Fragmentation profiles could also be used to identify the tissue of origin of the neoplasm. Combining fragmentation profiles with mutation-based cell-free DNA analyses detected 91% of patients with cancer.
Beyond mutational burden, methylation of ctDNA has recently emerged as a promising approach for cancer risk assessment and monitoring, especially for the detection of early tumours, including pancreatic cancer [95], or in colon cancer where a five-gene methylation panel can be used to compensate for the absence of patient-specific mutations to monitor tumour burden [96]. While challenging, this strategy has recently benefited from major technological advances [97], and could be proposed soon for assessing pharmacodynamics in clinical trials or when conventional ctDNA detection or imaging prove to be of limited use.

5. Exosome-Based Diagnostic for Pancreatic Cancer

Circulating tumour extracellular vesicles (EVs), including exosomes, are enriched with many bioactive molecules such as RNA, DNA, proteins, lipids and metabolites [98,99]. Once released from parental healthy or cancer cells, the cargoes reflect the status of that parent cell, and have the ability to relay signals between cells. For example, pancreatic cancer-derived exosomes loaded with tetraspanin 8 recruit proteins and mRNA cargo that can activate angiogenesis-related gene expression in neighbouring non-tumour endothelial cells [100]. EVs can also act at a distance. For example, Kupffer cells, the resident macrophages in the liver, have been shown to uptake pancreatic cancer derived exosomes containing the macrophage inhibitory factor (MIF). MIF promotes TGF-β (transforming growth factor beta) secretion by Kupffer cells, which in turns stimulates neighbouring hepatic stellate cells to secrete fibronectin, and creates local inflammation, considered to be the niche of pancreatic metastases [101]. Thus, due to their actual demonstrated activity, the detection of transported biomolecules protected from degradation by external nucleases or proteases presents an opportunity for both the diagnosis and prognosis of cancer.
There is currently no universal method for EV isolation/enrichment from body fluids, although recommendations from the International Society for Extracellular Vesicles have been first listed in 2014, and updated in 2018 [102]. Focusing on studies interested in PDAC, several methods have been described, all of them being complemented by PDAC-specific molecular characterisation of the enriched samples. For instance, enrichment by ultracentrifugation with or without sucrose gradients and ultrafiltration, or by kit-based precipitation have been reported (Table 3). More specifically, antigen-based exosome capture is also available using the CD63 exosome-specific tetraspanin in conjunction with microfluidic systems or magnetic beads. As EVs are released by any healthy or diseased cell, additional molecular characterisation of the obtained vesicles is required. Authors have been focusing on bioactive molecules carried by EVs such as nucleic acids and proteins.
As for other types of cancers [103] microRNA (miR) identification has been studied in the context of pancreatic cancer. As presented in Table 3, several, distinct miR signatures have been reported. From the testing of four individuals miRs (miR-17-5p, -21, -155, and -196a), miR-17-5p and -21 were shown to have a high diagnostic value, with a sensitivity and specificity between 72% and 95%. miR-155 and -196a were disregarded due to their low levels of expression in cancer exosomes [104]. Conversely, Xu et al. described an increased abundance of miR-196a, miR196b, or miR1246 exosomes in PDAC patients with areas under ROC curves (AUCs) ranking between 0.71 and 0.81. Other authors have found an increase in the expression of miR-191, miR-451a, and miR-21 in pancreatic cancer and IPMNs. Diagnostic accuracy was better than with CA-19.9 for early stage cancers, at approximately 80% [105]. Using a microarray approach on patient exosome samples, miR-1246, -4644, -3976, and -4306 were each found to be increased in PDAC samples [106]. Although theses authors did not report individual specificities and sensitivities, which were not 100% according to published figures, they did report that when all four miRs were combined they were detected in 9% of healthy controls (false positives) and in 80% of PDACs (20% of false negatives).
The prognostic value of miR quantification in exosomes was not evaluated by all authors. Unlike miR-21, the expression of miR-17-5p was correlated with tumour stages [104], although Goto et al. did find miR-21 to be prognostic for overall survival and chemo-resistance [105]. Interestingly, miR-451a was associated with patients with mural nodules in IPMNs [105], which is a sign of malignancy [114]. Quantifying miR-451a in exosome liquid biopsies could help in decision making for surgery of branch duct IPMNs. On the whole, a prognostic value has been found in five out of the 11 studies detailed in Table 3 (Figure 1).
It should be noted that circulating miR detection has superior sensitivity compared to ctDNA in the surgical setting; indeed, pre-operative plasma miR-21 was recently found as a sensitive biomarker and independent prognostic factor in patients with pancreatic cancer undergoing surgical resection [115]. Our group has participated in the demonstration that circulating miR sampled from different sources have biomarker value in preclinical models of PDAC and in patients. Briefly, we generated the first signature of salivary miR sampled from patients with locally advanced pancreatic tumours and found that selected salivary miRs, among them miR-23a, miR-23b, and miR-21, differentiate pancreatic cancer patients from patients with pancreatitis and matching healthy controls [115,116]. Interestingly, in mouse models of pancreatic cancer, the increase in salivary miR-23a, miR-23b and miR-21 levels precedes tumour burden detection by imaging [116]. In addition, in patients treated by gene therapy, we found that a panel of plasma miRs is predictive of response to therapy [117]. Since then, our group has partnered with physicists to develop novel nanodevices for the detection and quantification of candidate miRs from patients [118].
MiRs are not the only nucleic acid cargo of the exosomes. Unlike ctDNA, whose maximal size ranges from 150 to 170 base pairs [119], exosomes contain >10 kb fragments of double-stranded genomic DNA with detectable KRAS and p53 mutations when obtained from PDAC patients [120]. The difference in length observed between ctDNA and exoDNA is explained by the fact that DNA in exosomes is protected from nucleases. The relevance of exoDNA is actually a recent concept, and only a few studies have compared ctDNA and exoDNA diagnostic performances. Of note, specificity seemed to be impaired by the strength of the digital PCR-based positive signal in the detection of mutant KRAS in exoDNA in non-neoplastic patients examined in two recent reports. With mutation detection rates of 25% (3/12, [79]) and 7.4% (4/54, [109]) reported respectively. The KRAS mutation has already been described in a non-negligible proportion of plasma from healthy individuals (14/394, 3.5%, [121]) possibly reflecting a rare spontaneous somatic mutations. As for ctDNA, the high rates obtained in exoDNA might be linked to the highly sensitive detection method of KRAS mutants, which necessitate an interpretation considering the mutant allele frequency (MAF, [79]). The diagnostic performance of exoDNA was somewhat disappointing, since diagnostic accuracies ranked between 35% and 69% [79,109]. However, exoDNA showed relevance in PDAC management, as a correlation with progression free survival was detected in both studies, but this correlation was limited to patients with metastatic disease [106,113]. Despite its good prognostic value, exoDNA based on mutant KRAS detection using highly sensitive detection techniques, might not be suitable on its own for general population screening as it yields a high rate of false positives [79,109]. Additional biomarkers such as miRs or proteins should be included in the screening plan to improve the specificity.
Since protein cargo of EVs carry their biological function, diagnostic accuracy might be increased by protein marker identification. The membrane protein heparan sulfate proteoglycan glypican 1(GPC1) is overexpressed in several types of cancers, in particular in pancreatic primary tumours [122]. In 2015, a promising study quantified GPC1-positive exosomes in peripheral blood to diagnose PDAC and reported a sensitivity and a specificity of 100% [107]. Moreover, this circulating biomarker had strong prognostic value and could detect pre-neoplastic stages. In later studies, exosomal GPC1 alone failed to diagnose PDAC [108]. A signature with five exosomal surface proteins including GPC1 showed better sensitivity and specificity than each marker analysed separately (86% and 81%, [111]). Using an alternating current, electrokinetic (ACE) microarray chip that captures nano-particles including exosomes directly from plasma, followed by quantification of CD63 and GPC1, retrieved good but not perfect performance (sensitivity 99% specificity 82%) with false negatives and false positives [112]. Pulling down GPC1-expressing exosome did not increase diagnostic performance of miR-196a and miR-1246 [110]. Discrepancies in exosomal GPC1 validity in detecting PDAC might come from methodological differences. However, details on flow cytometry controls and settings published by Melo et al. raise questions on the validity of the published results. This might explain why no independent study has been able to confirm their results by using the same method. In addition to GPC1, the zinc transporter protein ZIP4 has recently been studied as an exosomal protein biomarker. The authors did not report diagnostic accuracy, but the AUC was 0.89. As was the case for GPC1, exosomal ZIP4 led to false-positive and false-negative results [113].
In conclusion, recent experimental applications utilising miRs, DNA, and exosome protein signatures, have not yet found a routine application for these techniques in the diagnosis of pancreatic cancer. Exosome isolation and characterisation is a major challenge in the field and is required to improve and standardise methods [102]. Another obstacle is the choice of the molecular signature. Exosomes are for instance very rich in differentially expressed miRs, as 119 miRs, with a 5-fold higher expression level detected in pancreatic cancer compared to healthy controls [106,123,124]. Among the numerous targets in each category, which have been selected by diverse strategies, only a few have been tested to date. Of the cited references here, it appears that combining miR-1246, miR-21, GPC1 and ZIP4 might be very relevant for the diagnosis of PDAC in particular.

6. Tumour-Educated Platelets

The team of Würdinger et al. has for many years explored how platelet RNA biomarker signatures can be altered by the presence of cancer [125] and reported an important role of these tumour-educated platelets (TEPs) as liquid biopsies in solid tumours (e.g., glioblastoma, non-small cell lung cancer, colorectal cancer, pancreatic cancer, breast cancer, and liver and bile duct carcinoma) [126]. Indeed, it has been previously shown that platelets interact with tumour cells and affect tumour growth and dissemination [127]. This interaction affects not only the expression of relevant genes in tumour cells, but also alters the RNA profile of blood platelets called TEPs. Interestingly, tumour-associated biomolecules can be transferred to platelets resulting in their education [125]. External stimuli (e.g., activation of platelet surface receptors and lipopolysaccharide-mediated platelet activation) induce specific splicing of pre-mRNAs in TEPs. Thus, RNA sequencing (thromboSeq technique) performed on 228 blood platelet samples from patients with different tumour types including pancreatic cancer showed that the location of the primary tumour was correctly identified with 71% accuracy [126]. Moreover, TEPs mRNA profiles could distinguish mutant KRAS, EGFR, or PIK3CA tumours, which is a crucial application in oncology. In conclusion, the ability of TEPs to identify precisely the location of the primary tumour as well as its molecular composition opens a new avenue to use liquid biopsies for cancer diagnostic. An important step in the future will be to demonstrate the clinical utility of TEPs as liquid biopsy biosources in large prospective clinical trials.

7. Blood-Based Protein and Metabolite Markers

Recent studies have addressed the role of circulating proteins for the diagnosis and the prognosis of PDAC. In most of these studies, the candidate biomarker is compared with CA 19-9. Two studies investigated the diagnostic value of thrombospondin-2 (THBS2), which is a disulfide-linked homotrimetric glycoprotein that mediates cell-to-cell and cell-to-matrix interactions [128,129]. The levels of THBS2 were compared in patients with PDAC, as compared to patients with benign pancreatic diseases. While the diagnostic performance of the THBS2 (ELISA) plasma assay is similar to that of CA19-9 (area under curve of the ROC 0.73 to 0.94) (AUC), the combination of the two assays significantly improves these performances (AUC: 0.90 to 0.97). A team from Texas University MD Anderson Cancer Center recently demonstrated that tissue metalloproteinase inhibitor-1 (TIMP1) and the Leucine-rich alpha-2-glycoprotein 1 (LRG12) expression detection shows significant performance for the diagnosis of early stage PDAC (AUC: 0.95), when associated with CA 19-9 detection [130]. In a similar study, they found that 5 metabolites show interesting diagnostic performance, either alone (AUC 0.72–0.84), or in combination (0.82–0.95). Combining this metabolomics signature with TIMP1, LRG1, and CA 19-9 detection gives a remarkable AUC of 0.92 [131]. Another team studied the diagnostic value of a combination of nine metabolites associated with the CA 19-9 assay. Of these nine candidates, five were able to discriminate PDAC and chronic pancreatitis with diagnostic values up to 0.95 [132]. Finally, a recent study revealed that high circulating levels of inhibitory immune checkpoints (such as PDL-1, BTLA, BTN3A1, and BTN3A) correlate with shorter survival [133].

8. Conclusions and Future Directions

The aim of liquid biopsies is to increase the negative predictive value of the gold EUS-FNA cytopathology standard but also to ensure the rapid diagnosis of PDAC in order to facilitate its early management. Other aims are to hopefully identify tools and conditions for a better evaluation of the prognosis as well as to better monitor PDAC patients, before and after surgical resection and in addition to allow prediction of relapses. However, there are still some unknowns regarding the use of liquid biopsies in the current practice for PDAC. Indeed, there is an obvious heterogeneity of the methods, regarding either the type of biomarker source “cells, DNA or exosomes” and the molecular targets used to identify them. The idea would be to get combined information as an index or an algorithm to get a more precise picture of cancer progression (e.g., quantitative and qualitative). On the other hand, standardisation is required before (or after) establishing the real place of liquid biopsies in the management of PDAC. With regard to diagnosis, the results are more convincing in the case of locally advanced and/or metastatic cancers, considering the size of tumour mass and diffusion. Nevertheless, the improvement of detection methods (such as the dPCR for the detection of the KRAS oncogene mutation) and the combination of liquid biopsy types (example of ExoDNA) should allow us to obtain a diagnostic yield that fits the current clinical practice. Thus, more emphasis on technical validation is required, and projects such as the European CANCER-ID, European Liquid Biopsy Academy (ELBA) and European Liquid Biopsy Society (ELBS) networks or the U.S.-based BloodPAC have been initiated to meet this challenge. These consortia combine the expertise of academic and industry partners and will hopefully lead to the development of robust liquid biopsy assays and inform the design of the trials needed to prove the clinical utility of liquid biopsy testing. These societies and consortia attempt also to find an agreement on the best way to collect, store and handle this type of sampling and analysis in such a way as to not only unify but also centralize it at expert centres.
Beside standardization of the methods, more studies are needed including a larger number of patients. Indeed, from a statistical point of view, most published studies have included between 50 and 75 patients with PDAC and the number of controls is not higher. It is obvious that any robust assessment of the sensitivity and specificity of a diagnostic method requires a sufficient number of patients, taking into account the heterogeneity of the disease (i.e., metastatic, locally advanced, borderline). The same is true for prognosis, which is certainly appreciable for all types of patients by measuring overall survival, but the evaluation of progression-free survival or recurrence after surgery is just as important. For this purpose, subgroups with a sufficient number of patients must also be studied.
Recent translational discoveries pinpoint that PDAC tumours are highly heterogeneous, at the molecular and cellular levels that can account at least in part to PDAC extreme resistance to treatment. This heterogeneity reflects the difficulty not only of early diagnosis but also of selecting reliable biomarkers. There are many markers and this is found in the field of liquid biopsies. Indeed, the only example of exosomes attests to this variety and heterogeneity since proteins, DNA, miRs coexist. This reflects the value of combining several methods. Recent examples have been given by the isolation of circulating exosomes carrying ctDNA or proteins of interest [79,99].
The role of liquid biopsies to assess PDAC prognoses is also evident, but in the absence of a significant therapeutic arsenal, the current therapeutic attitude for locally advanced or metastatic cancers will continue to be used. However, this is not to imply that we should sit twiddling our thumbs; our efforts must be doubled to identify biomarkers but also nanodevices for detection in the likely event that progress will be made in the therapeutic management of these advanced forms of the disease.
On the other hand, for patients with a resectable tumours and/or without obvious metastases, the contribution of liquid biopsies for the diagnosis of small metastases or the prediction of an early relapse after surgery would be of great benefit. Indeed, the surgical decision, the neoadjuvant treatment, and the intensification of adjuvant treatment could all be modified and decided on this basis.
In addition, these new non-invasive methods which lend themselves to frequent sampling, could certainly benefit from a combination of several approaches by concomitantly detecting several circulating tumour elements. A representative example is the exosomes that are carriers of tumour DNA; these might indeed be representative of tumour spread and metastasis. This approach would allow the evaluation and monitoring of the “distant activity” of PDAC. In addition, the use of NGS or miR profiling containing specific “molecular signatures” that have been already characterised in pilot studies (as detailed in the present review) may also improve the sensitivity of liquid biopsies in PDAC and could be integrated as a tool for personalised medicine.
The improvement of technology for detection and isolation of CTCs and exosomes will certainly be of importance, as well as pilot studies comparing blood samples near the tumour (portal vein in the case of PDAC) and peripheral blood. In addition, isolation of CTCs will be also be a great model for in vivo studies of tumour progression and response to treatment.
In conclusion, all published studies suggest that real-time liquid biopsy is a highly promising clinical tool for the non-invasive diagnosis and prognosis of PDAC in the future. Combination of methods will certainly be the key point of these promising modern methods of diagnosis.

Author Contributions

Conceptualisation E.B., S.D., C.A.-P. and L.B.; Collecting data: E.B. and C.M.; Writing and draft preparation: E.B., S.D., C.A.-P. and L.B.; Review and editing: S.D., C.A.-P., L.C., F.M., P.C. and L.B.; Approving the final version of the manuscript: E.B., S.D., C.A.-P., L.B., C.M., L.C., P.C., F.M.

Funding

This review article received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. 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. [Google Scholar] [CrossRef]
  2. Bouvier, A.-M.; Uhry, Z.; Jooste, V.; Drouillard, A.; Remontet, L.; Launoy, G.; Leone, N. French Network of Cancer Registries (FRANCIM) Focus on an unusual rise in pancreatic cancer incidence in France. Int. J. Epidemiol. 2017, 46, 1764–1772. [Google Scholar] [CrossRef]
  3. Ryan, D.P.; Hong, T.S.; Bardeesy, N. Pancreatic adenocarcinoma. N. Engl. J. Med. 2014, 371, 1039–1049. [Google Scholar] [CrossRef]
  4. Neoptolemos, J.P.; Kleeff, J.; Michl, P.; Costello, E.; Greenhalf, W.; Palmer, D.H. Therapeutic developments in pancreatic cancer: Current and future perspectives. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 333–348. [Google Scholar] [CrossRef]
  5. Conroy, T.; Hammel, P.; Hebbar, M.; Ben Abdelghani, M.; Wei, A.C.; Raoul, J.-L.; Choné, L.; Francois, E.; Artru, P.; Biagi, J.J.; et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N. Engl. J. Med. 2018, 379, 2395–2406. [Google Scholar] [CrossRef]
  6. Buscail, L. Commentary: Pancreatic cancer: Is the worst to come? Int. J. Epidemiol. 2017, 46, 1774–1775. [Google Scholar] [CrossRef]
  7. Imamura, T.; Komatsu, S.; Ichikawa, D.; Kawaguchi, T.; Miyamae, M.; Okajima, W.; Ohashi, T.; Arita, T.; Konishi, H.; Shiozaki, A.; et al. Liquid biopsy in patients with pancreatic cancer: Circulating tumor cells and cell-free nucleic acids. World J. Gastroenterol. 2016, 22, 5627–5641. [Google Scholar] [CrossRef]
  8. Zhang, R.; Peng, R.; Li, Z.; Gao, P.; Jia, S.; Yang, X.; Ding, J.; Han, Y.; Xie, J.; Li, J. Synthetic Circulating Cell-free DNA as Quality Control Materials for Somatic Mutation Detection in Liquid Biopsy for Cancer. Clin. Chem. 2017, 63, 1465–1475. [Google Scholar] [CrossRef]
  9. Pantel, K.; Alix-Panabières, C. Liquid biopsy and minimal residual disease—Latest advances and implications for cure. Nat. Rev. Clin. Oncol. 2019. [Google Scholar] [CrossRef]
  10. Alix-Panabières, C.; Pantel, K. Circulating tumor cells: Liquid biopsy of cancer. Clin. Chem. 2013, 59, 110–118. [Google Scholar] [CrossRef]
  11. Chudasama, D.; Katopodis, P.; Stone, N.; Haskell, J.; Sheridan, H.; Gardner, B.; Urnovitz, H.; Schuetz, E.; Beck, J.; Hall, M.; et al. Liquid Biopsies in Lung Cancer: Four Emerging Technologies and Potential Clinical Applications. Cancers 2019, 11, 311. [Google Scholar] [CrossRef]
  12. Stefanovic, S.; Deutsch, T.M.; Wirtz, R.; Hartkopf, A.; Sinn, P.; Schuetz, F.; Sohn, C.; Bohlmann, M.K.; Sütterlin, M.; Schneeweiss, A.; et al. Molecular Subtype Conversion between Primary and Metastatic Breast Cancer Corresponding to the Dynamics of Apoptotic and Intact Circulating Tumor Cells. Cancers 2019, 11, 342. [Google Scholar] [CrossRef]
  13. Zhang, L.; Sanagapalli, S.; Stoita, A. Challenges in diagnosis of pancreatic cancer. World J. Gastroenterol. 2018, 24, 2047–2060. [Google Scholar] [CrossRef]
  14. Lee, E.S.; Lee, J.M. Imaging diagnosis of pancreatic cancer: A state-of-the-art review. World J. Gastroenterol. 2014, 20, 7864–7877. [Google Scholar] [CrossRef]
  15. Puli, S.R.; Bechtold, M.L.; Buxbaum, J.L.; Eloubeidi, M.A. How good is endoscopic ultrasound-guided fine-needle aspiration in diagnosing the correct etiology for a solid pancreatic mass?: A meta-analysis and systematic review. Pancreas 2013, 42, 20–26. [Google Scholar] [CrossRef]
  16. Buscail, L.; Faure, P.; Bournet, B.; Selves, J.; Escourrou, J. Interventional endoscopic ultrasound in pancreatic diseases. Pancreatology 2006, 6, 7–16. [Google Scholar] [CrossRef]
  17. Savides, T.J.; Donohue, M.; Hunt, G.; Al-Haddad, M.; Aslanian, H.; Ben-Menachem, T.; Chen, V.K.; Coyle, W.; Deutsch, J.; DeWitt, J.; et al. EUS-guided FNA diagnostic yield of malignancy in solid pancreatic masses: A benchmark for quality performance measurement. Gastrointest. Endosc. 2007, 66, 277–282. [Google Scholar] [CrossRef]
  18. Yoshinaga, S.; Suzuki, H.; Oda, I.; Saito, Y. Role of endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) for diagnosis of solid pancreatic masses. Dig. Endosc. 2011, 23 (Suppl. 1), 29–33. [Google Scholar] [CrossRef]
  19. Bournet, B.; Buscail, C.; Muscari, F.; Cordelier, P.; Buscail, L. Targeting KRAS for diagnosis, prognosis, and treatment of pancreatic cancer: Hopes and realities. Eur. J. Cancer 2016, 54, 75–83. [Google Scholar] [CrossRef]
  20. Fuccio, L.; Hassan, C.; Laterza, L.; Correale, L.; Pagano, N.; Bocus, P.; Fabbri, C.; Maimone, A.; Cennamo, V.; Repici, A.; et al. The role of K-ras gene mutation analysis in EUS-guided FNA cytology specimens for the differential diagnosis of pancreatic solid masses: A meta-analysis of prospective studies. Gastrointest. Endosc. 2013, 78, 596–608. [Google Scholar] [CrossRef]
  21. Fusaroli, P.; Spada, A.; Mancino, M.G.; Caletti, G. Contrast harmonic echo-endoscopic ultrasound improves accuracy in diagnosis of solid pancreatic masses. Clin. Gastroenterol. Hepatol. 2010, 8, 629–634.e1–2. [Google Scholar] [CrossRef]
  22. Sanjeevi, S.; Ivanics, T.; Lundell, L.; Kartalis, N.; Andrén-Sandberg, Å.; Blomberg, J.; Del Chiaro, M.; Ansorge, C. Impact of delay between imaging and treatment in patients with potentially curable pancreatic cancer. Br. J. Surg. 2016, 103, 267–275. [Google Scholar] [CrossRef]
  23. Bournet, B.; Pointreau, A.; Delpu, Y.; Selves, J.; Torrisani, J.; Buscail, L.; Cordelier, P. Molecular endoscopic ultrasound for diagnosis of pancreatic cancer. Cancers 2011, 3, 872–882. [Google Scholar] [CrossRef]
  24. Bournet, B.; Selves, J.; Grand, D.; Danjoux, M.; Hanoun, N.; Cordelier, P.; Buscail, L. Endoscopic ultrasound-guided fine-needle aspiration biopsy coupled with a KRAS mutation assay using allelic discrimination improves the diagnosis of pancreatic cancer. J. Clin. Gastroenterol. 2015, 49, 50–56. [Google Scholar] [CrossRef]
  25. Trisolini, E.; Armellini, E.; Paganotti, A.; Veggiani, C.; Bozzola, C.; Frattini, M.; Pizio, C.; Mancuso, G.; Andorno, S.; Boldorini, R. KRAS mutation testing on all non-malignant diagnosis of pancreatic endoscopic ultrasound-guided fine-needle aspiration biopsies improves diagnostic accuracy. Pathology 2017, 49, 379–386. [Google Scholar] [CrossRef]
  26. Sekita-Hatakeyama, Y.; Nishikawa, T.; Takeuchi, M.; Morita, K.; Takeda, M.; Hatakeyama, K.; Nakai, T.; Uchiyama, T.; Itami, H.; Fujii, T.; et al. K-ras mutation analysis of residual liquid-based cytology specimens from endoscopic ultrasound-guided fine needle aspiration improves cell block diagnosis of pancreatic ductal adenocarcinoma. PLoS ONE 2018, 13, e0193692. [Google Scholar] [CrossRef]
  27. Alix-Panabières, C.; Pantel, K. Challenges in circulating tumour cell research. Nat. Rev. Cancer 2014, 14, 623–631. [Google Scholar] [CrossRef]
  28. Samandari, M.; Julia, M.G.; Rice, A.; Chronopoulos, A.; Del Rio Hernandez, A.E. Liquid biopsies for management of pancreatic cancer. Transl. Res. 2018, 201, 98–127. [Google Scholar] [CrossRef]
  29. 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. [Google Scholar] [CrossRef]
  30. Soeth, E.; Grigoleit, U.; Moellmann, B.; Röder, C.; Schniewind, B.; Kremer, B.; Kalthoff, H.; Vogel, I. Detection of tumor cell dissemination in pancreatic ductal carcinoma patients by CK 20 RT-PCR indicates poor survival. J. Cancer Res. Clin. Oncol. 2005, 131, 669–676. [Google Scholar] [CrossRef]
  31. Sergeant, G.; Roskams, T.; van Pelt, J.; Houtmeyers, F.; Aerts, R.; Topal, B. Perioperative cancer cell dissemination detected with a real-time RT-PCR assay for EpCAM is not associated with worse prognosis in pancreatic ductal adenocarcinoma. BMC Cancer 2011, 11, 47. [Google Scholar] [CrossRef]
  32. Zhou, J.; Hu, L.; Yu, Z.; Zheng, J.; Yang, D.; Bouvet, M.; Hoffman, R.M. Marker expression in circulating cancer cells of pancreatic cancer patients. J. Surg. Res. 2011, 171, 631–636. [Google Scholar] [CrossRef]
  33. Ren, C.; Han, C.; Zhang, J.; He, P.; Wang, D.; Wang, B.; Zhao, P.; Zhao, X. Detection of apoptotic circulating tumor cells in advanced pancreatic cancer following 5-fluorouracil chemotherapy. Cancer Biol. Ther. 2011, 12, 700–706. [Google Scholar] [CrossRef] [Green Version]
  34. Khoja, L.; Backen, A.; Sloane, R.; Menasce, L.; Ryder, D.; Krebs, M.; Board, R.; Clack, G.; Hughes, A.; Blackhall, F.; et al. A pilot study to explore circulating tumour cells in pancreatic cancer as a novel biomarker. Br. J. Cancer 2012, 106, 508–516. [Google Scholar] [CrossRef] [PubMed]
  35. Ankeny, J.S.; Court, C.M.; Hou, S.; Li, Q.; Song, M.; Wu, D.; Chen, J.F.; Lee, T.; Lin, M.; Sho, S.; et al. Circulating tumour cells as a biomarker for diagnosis and staging in pancreatic cancer. Br. J. Cancer 2016, 114, 1367–1375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Dotan, E.; Alpaugh, R.K.; Ruth, K.; Negin, B.P.; Denlinger, C.S.; Hall, M.J.; Astsaturov, I.; McAleer, C.; Fittipaldi, P.; Thrash-Bingham, C.; et al. Prognostic Significance of MUC-1 in Circulating Tumor Cells in Patients With Metastatic Pancreatic Adenocarcinoma. Pancreas 2016, 45, 1131–1135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Poruk, K.E.; Blackford, A.L.; Weiss, M.J.; Cameron, J.L.; He, J.; Goggins, M.; Rasheed, Z.A.; Wolfgang, C.L.; Wood, L.D. Circulating Tumor Cells Expressing Markers of Tumor-Initiating Cells Predict Poor Survival and Cancer Recurrence in Patients with Pancreatic Ductal Adenocarcinoma. Clin. Cancer Res. 2017, 23, 2681–2690. [Google Scholar] [CrossRef] [PubMed]
  38. Kulemann, B.; Rösch, S.; Seifert, S.; Timme, S.; Bronsert, P.; Seifert, G.; Martini, V.; Kuvendjiska, J.; Glatz, T.; Hussung, S.; et al. Pancreatic cancer: Circulating Tumor Cells and Primary Tumors show Heterogeneous KRAS Mutations. Sci. Rep. 2017, 7, 4510. [Google Scholar] [CrossRef] [PubMed]
  39. Okubo, K.; Uenosono, Y.; Arigami, T.; Mataki, Y.; Matsushita, D.; Yanagita, S.; Kurahara, H.; Sakoda, M.; Kijima, Y.; Maemura, K.; et al. Clinical impact of circulating tumor cells and therapy response in pancreatic cancer. Eur. J. Surg. Oncol. 2017, 43, 1050–1055. [Google Scholar] [CrossRef]
  40. Court, C.M.; Ankeny, J.S.; Sho, S.; Winograd, P.; Hou, S.; Song, M.; Wainberg, Z.A.; Girgis, M.D.; Graeber, T.G.; Agopian, V.G.; et al. Circulating Tumor Cells Predict Occult Metastatic Disease and Prognosis in Pancreatic Cancer. Ann. Surg. Oncol. 2018, 25, 1000–1008. [Google Scholar] [CrossRef]
  41. Effenberger, K.E.; Schroeder, C.; Hanssen, A.; Wolter, S.; Eulenburg, C.; Tachezy, M.; Gebauer, F.; Izbicki, J.R.; Pantel, K.; Bockhorn, M. Improved Risk Stratification by Circulating Tumor Cell Counts in Pancreatic Cancer. Clin. Cancer Res. 2018, 24, 2844–2850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Hugenschmidt, H.; Labori, K.J.; Brunborg, C.; Verbeke, C.S.; Seeberg, L.T.; Schirmer, C.B.; Renolen, A.; Borgen, E.F.; Naume, B.; Wiedswang, G. Circulating Tumor Cells are an Independent Predictor of Shorter Survival in Patients Undergoing Resection for Pancreatic and Periampullary Adenocarcinoma. Ann. Surg. 2018. [Google Scholar] [CrossRef] [PubMed]
  43. Sefrioui, D.; Blanchard, F.; Toure, E.; Basile, P.; Beaussire, L.; Dolfus, C.; Perdrix, A.; Paresy, M.; Antonietti, M.; Iwanicki-Caron, I.; et al. Diagnostic value of CA19.9, circulating tumour DNA and circulating tumour cells in patients with solid pancreatic tumours. Br. J. Cancer 2017, 117, 1017–1025. [Google Scholar] [CrossRef] [PubMed]
  44. Varillas, J.I.; Zhang, J.; Chen, K.; Barnes, I.I.; Liu, C.; George, T.J.; Fan, Z.H. Microfluidic Isolation of Circulating Tumor Cells and Cancer Stem-Like Cells from Patients with Pancreatic Ductal Adenocarcinoma. Theranostics 2019, 9, 1417–1425. [Google Scholar] [CrossRef] [PubMed]
  45. Wei, T.; Zhang, X.; Zhang, Q.; Yang, J.; Chen, Q.; Wang, J.; Li, X.; Chen, J.; Ma, T.; Li, G.; et al. Vimentin-positive circulating tumor cells as a biomarker for diagnosis and treatment monitoring in patients with pancreatic cancer. Cancer Lett. 2019, 452, 237–243. [Google Scholar] [CrossRef] [PubMed]
  46. Tam, W.L.; Weinberg, R.A. The epigenetics of epithelial-mesenchymal plasticity in cancer. Nat. Med. 2013, 19, 1438–1449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Chaffer, C.L.; Weinberg, R.A. A perspective on cancer cell metastasis. Science 2011, 331, 1559–1564. [Google Scholar] [CrossRef] [PubMed]
  48. Alix-Panabières, C.; Mader, S.; Pantel, K. Epithelial-mesenchymal plasticity in circulating tumor cells. J. Mol. Med. 2017, 95, 133–142. [Google Scholar] [CrossRef]
  49. Vona, G.; Sabile, A.; Louha, M.; Sitruk, V.; Romana, S.; Schütze, K.; Capron, F.; Franco, D.; Pazzagli, M.; Vekemans, M.; et al. Isolation by size of epithelial tumor cells: A new method for the immunomorphological and molecular characterization of circulatingtumor cells. Am. J. Pathol. 2000, 156, 57–63. [Google Scholar] [CrossRef]
  50. Rosenbaum, M.W.; Cauley, C.E.; Kulemann, B.; Liss, A.S.; Castillo, C.F.-D.; Warshaw, A.L.; Lillemoe, K.D.; Thayer, S.P.; Pitman, M.B. Cytologic characteristics of circulating epithelioid cells in pancreatic disease. Cancer Cytopathol. 2017, 125, 332–340. [Google Scholar] [CrossRef] [Green Version]
  51. Pantel, K.; Denève, E.; Nocca, D.; Coffy, A.; Vendrell, J.-P.; Maudelonde, T.; Riethdorf, S.; Alix-Panabières, C. Circulating epithelial cells in patients with benign colon diseases. Clin. Chem. 2012, 58, 936–940. [Google Scholar] [CrossRef]
  52. Bidard, F.C.; Huguet, F.; Louvet, C.; Mineur, L.; Bouché, O.; Chibaudel, B.; Artru, P.; Desseigne, F.; Bachet, J.B.; Mathiot, C.; et al. Circulating tumor cells in locally advanced pancreatic adenocarcinoma: The ancillary CirCe 07 study to the LAP 07 trial. Ann. Oncol. 2013, 24, 2057–2061. [Google Scholar] [CrossRef]
  53. Chin, R.-I.; Chen, K.; Usmani, A.; Chua, C.; Harris, P.K.; Binkley, M.S.; Azad, T.D.; Dudley, J.C.; Chaudhuri, A.A. Detection of Solid Tumor Molecular Residual Disease (MRD) Using Circulating Tumor DNA (ctDNA). Mol. Diagn. Ther. 2019, 23, 311–331. [Google Scholar] [CrossRef]
  54. Lewis, A.R.; Valle, J.W.; McNamara, M.G. Pancreatic cancer: Are “liquid biopsies” ready for prime-time? World J. Gastroenterol. 2016, 22, 7175–7185. [Google Scholar] [CrossRef]
  55. Castells, A.; Puig, P.; Móra, J.; Boadas, J.; Boix, L.; Urgell, E.; Solé, M.; Capellà, G.; Lluís, F.; Fernández-Cruz, L.; et al. 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. [Google Scholar] [CrossRef]
  56. Maire, F.; Micard, S.; Hammel, P.; Voitot, H.; Lévy, P.; Cugnenc, P.-H.; Ruszniewski, P.; Puig, P.L. 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. [Google Scholar] [CrossRef]
  57. Däbritz, J.; Preston, R.; Hänfler, J.; Oettle, H. Follow-up study of K-ras mutations in the plasma of patients with pancreatic cancer: Correlation with clinical features and carbohydrate antigen 19-9. Pancreas 2009, 38, 534–541. [Google Scholar] [CrossRef]
  58. Chen, H.; Tu, H.; Meng, Z.Q.; Chen, Z.; Wang, P.; Liu, L.M. K-ras mutational status predicts poor prognosis in unresectable pancreatic cancer. Eur. J. Surg. Oncol. 2010, 36, 657–662. [Google Scholar] [CrossRef]
  59. Wu, J.; Zhou, Y.; Zhang, C.-Y.; Song, B.-B.; Wang, B.-L.; Pan, B.-S.; Lou, W.-H.; Guo, W. Co-amplification at lower denaturation-temperature PCR combined with unlabled-probe high-resolution melting to detect KRAS codon 12 and 13 mutations in plasma-circulating DNA of pancreatic adenocarcinoma cases. Asian Pac. J. Cancer Prev. 2014, 15, 10647–10652. [Google Scholar] [CrossRef]
  60. Semrad, T.; Barzi, A.; Lenz, H.-J.; Hutchins, I.M.; Kim, E.J.; Gong, I.-Y.; Tanaka, M.; Beckett, L.; Holland, W.; Burich, R.A.; et al. Pharmacodynamic separation of gemcitabine and erlotinib in locally advanced or metastatic pancreatic cancer: Therapeutic and biomarker results. Int. J. Clin. Oncol. 2015, 20, 518–524. [Google Scholar] [CrossRef]
  61. Sausen, M.; Phallen, J.; Adleff, V.; Jones, S.; Leary, R.J.; Barrett, M.T.; Anagnostou, V.; Parpart-Li, S.; Murphy, D.; Kay Li, Q.; et al. Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients. Nat. Commun. 2015, 6, 7686. [Google Scholar] [CrossRef]
  62. Earl, J.; Garcia-Nieto, S.; Martinez-Avila, J.C.; Montans, J.; Sanjuanbenito, A.; Rodríguez-Garrote, M.; Lisa, E.; Mendía, E.; Lobo, E.; Malats, N.; et al. 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. [Google Scholar] [CrossRef]
  63. Singh, N.; Gupta, S.; Pandey, R.M.; Chauhan, S.S.; Saraya, A. High levels of cell-free circulating nucleic acids in pancreatic cancer are associated with vascular encasement, metastasis and poor survival. Cancer Investig. 2015, 33, 78–85. [Google Scholar] [CrossRef]
  64. 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. [Google Scholar] [CrossRef]
  65. Takai, E.; Totoki, Y.; Nakamura, H.; Morizane, C.; Nara, S.; Hama, N.; Suzuki, M.; Furukawa, E.; Kato, M.; Hayashi, H.; et al. Clinical utility of circulating tumor DNA for molecular assessment in pancreatic cancer. Sci. Rep. 2015, 5, 18425. [Google Scholar] [CrossRef]
  66. Adamo, P.; Cowley, C.M.; Neal, C.P.; Mistry, V.; Page, K.; Dennison, A.R.; Isherwood, J.; Hastings, R.; Luo, J.; Moore, D.A.; et al. Profiling tumour heterogeneity through circulating tumour DNA in patients with pancreatic cancer. Oncotarget 2017, 8, 87221–87233. [Google Scholar] [CrossRef] [Green Version]
  67. Ako, S.; Nouso, K.; Kinugasa, H.; Dohi, C.; Matushita, H.; Mizukawa, S.; Muro, S.; Akimoto, Y.; Uchida, D.; Tomoda, T.; et al. Utility of serum DNA as a marker for KRAS mutations in pancreatic cancer tissue. Pancreatology 2017, 17, 285–290. [Google Scholar] [CrossRef]
  68. Cheng, H.; Liu, C.; Jiang, J.; Luo, G.; Lu, Y.; Jin, K.; Guo, M.; Zhang, Z.; Xu, J.; Liu, L.; et al. Analysis of ctDNA to predict prognosis and monitor treatment responses in metastatic pancreatic cancer patients. Int. J. Cancer 2017, 140, 2344–2350. [Google Scholar] [CrossRef]
  69. Pietrasz, D.; Pécuchet, N.; Garlan, F.; Didelot, A.; Dubreuil, O.; Doat, S.; Imbert-Bismut, F.; Karoui, M.; Vaillant, J.-C.; Taly, V.; et al. Plasma Circulating Tumor DNA in Pancreatic Cancer Patients Is a Prognostic Marker. Clin. Cancer Res. 2017, 23, 116–123. [Google Scholar] [CrossRef]
  70. Van Laethem, J.-L.; Riess, H.; Jassem, J.; Haas, M.; Martens, U.M.; Weekes, C.; Peeters, M.; Ross, P.; Bridgewater, J.; Melichar, B.; et al. Phase I/II Study of Refametinib (BAY 86-9766) in Combination with Gemcitabine in Advanced Pancreatic cancer. Target. Oncol. 2017, 12, 97–109. [Google Scholar] [CrossRef]
  71. Hadano, N.; Murakami, Y.; Uemura, K.; Hashimoto, Y.; Kondo, N.; Nakagawa, N.; Sueda, T.; Hiyama, E. Prognostic value of circulating tumour DNA in patients undergoing curative resection for pancreatic cancer. Br. J. Cancer 2016, 115, 59–65. [Google Scholar] [CrossRef]
  72. Del Re, M.; Vivaldi, C.; Rofi, E.; Vasile, E.; Miccoli, M.; Caparello, C.; d’Arienzo, P.D.; Fornaro, L.; Falcone, A.; Danesi, R. Early changes in plasma DNA levels of mutant KRAS as a sensitive marker of response to chemotherapy in pancreatic cancer. Sci. Rep. 2017, 7, 7931. [Google Scholar] [CrossRef]
  73. Cohen, J.D.; Javed, A.A.; Thoburn, C.; Wong, F.; Tie, J.; Gibbs, P.; Schmidt, C.M.; Yip-Schneider, M.T.; Allen, P.J.; Schattner, M.; et al. Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers. Proc. Natl. Acad. Sci. USA 2017, 114, 10202–10207. [Google Scholar] [CrossRef] [Green Version]
  74. Pishvaian, M.J.; Joseph Bender, R.; Matrisian, L.M.; Rahib, L.; Hendifar, A.; Hoos, W.A.; Mikhail, S.; Chung, V.; Picozzi, V.; Heartwell, C.; et al. A pilot study evaluating concordance between blood-based and patient-matched tumor molecular testing within pancreatic cancer patients participating in the Know Your Tumor (KYT) initiative. Oncotarget 2017, 8, 83446–83456. [Google Scholar] [CrossRef]
  75. Kim, M.K.; Woo, S.M.; Park, B.; Yoon, K.-A.; Kim, Y.-H.; Joo, J.; Lee, W.J.; Han, S.-S.; Park, S.-J.; Kong, S.-Y. Prognostic Implications of Multiplex Detection of KRAS Mutations in Cell-Free DNA from Patients with Pancreatic Ductal Adenocarcinoma. Clin. Chem. 2018, 64, 726–734. [Google Scholar] [CrossRef] [Green Version]
  76. Nakano, Y.; Kitago, M.; Matsuda, S.; Nakamura, Y.; Fujita, Y.; Imai, S.; Shinoda, M.; Yagi, H.; Abe, Y.; Hibi, T.; et al. KRAS mutations in cell-free DNA from preoperative and postoperative sera as a pancreatic cancer marker: A retrospective study. Br. J. Cancer 2018, 118, 662–669. [Google Scholar] [CrossRef]
  77. Lin, M.; Alnaggar, M.; Liang, S.; Chen, J.; Xu, K.; Dong, S.; Du, D.; Niu, L. Circulating Tumor DNA as a Sensitive Marker in Patients Undergoing Irreversible Electroporation for Pancreatic Cancer. Cell. Physiol. Biochem. 2018, 47, 1556–1564. [Google Scholar] [CrossRef]
  78. Gall, T.M.H.; Belete, S.; Khanderia, E.; Frampton, A.E.; Jiao, L.R. Circulating Tumor Cells and Cell-Free DNA in Pancreatic Ductal Adenocarcinoma. Am. J. Pathol. 2019, 189, 71–81. [Google Scholar] [CrossRef] [Green Version]
  79. Bernard, V.; Kim, D.U.; San Lucas, F.A.; Castillo, J.; Allenson, K.; Mulu, F.C.; Stephens, B.M.; Huang, J.; Semaan, A.; Guerrero, P.A.; et al. Circulating Nucleic Acids Are Associated With Outcomes of Patients with Pancreatic Cancer. Gastroenterology 2019, 156, 108–118.e4. [Google Scholar] [CrossRef]
  80. Anderson, S.M. Laboratory methods for KRAS mutation analysis. Expert Rev. Mol. Diagn. 2011, 11, 635–642. [Google Scholar] [CrossRef]
  81. Pritchard, C.C.; Akagi, L.; Reddy, P.L.; Joseph, L.; Tait, J.F. COLD-PCR enhanced melting curve analysis improves diagnostic accuracy for KRAS mutations in colorectal carcinoma. BMC Clin. Pathol. 2010, 10, 6. [Google Scholar] [CrossRef]
  82. Oliner, K.; Juan, T.; Suggs, S.; Wolf, M.; Sarosi, I.; Freeman, D.J.; Gyuris, T.; Baron, W.; Bakker, A.; Parker, A.; et al. A comparability study of 5 commercial KRAS tests. Diagn. Pathol. 2010, 5, 23. [Google Scholar] [CrossRef]
  83. Boulaiz, H.; Ramos, M.C.; Griñán-Lisón, C.; García-Rubiño, M.E.; Vicente, F.; Marchal, J.A. What’s new in the diagnosis of pancreatic cancer: A patent review (2011-present). Expert Opin. Ther. Pat. 2017, 27, 1319–1328. [Google Scholar] [CrossRef]
  84. Sho, S.; Court, C.M.; Kim, S.; Braxton, D.R.; Hou, S.; Muthusamy, V.R.; Watson, R.R.; Sedarat, A.; Tseng, H.-R.; Tomlinson, J.S. Digital PCR Improves Mutation Analysis in Pancreas Fine Needle Aspiration Biopsy Specimens. PLoS ONE 2017, 12, e0170897. [Google Scholar] [CrossRef]
  85. Henriksen, S.D.; Madsen, P.H.; Larsen, A.C.; Johansen, M.B.; Pedersen, I.S.; Krarup, H.; Thorlacius-Ussing, O. Cell-free DNA promoter hypermethylation in plasma as a predictive marker for survival of patients with pancreatic adenocarcinoma. Oncotarget 2017, 8, 93942–93956. [Google Scholar] [CrossRef]
  86. Kawesha, A.; Ghaneh, P.; Andrén-Sandberg, A.; Ograed, D.; Skar, R.; Dawiskiba, S.; Evans, J.D.; Campbell, F.; Lemoine, N.; Neoptolemos, J.P. K-ras oncogene subtype mutations are associated with survival but not expression of p53, p16(INK4A), p21(WAF-1), cyclin D1, erbB-2 and erbB-3 in resected pancreatic ductal adenocarcinoma. Int. J. Cancer 2000, 89, 469–474. [Google Scholar] [CrossRef]
  87. Kim, S.T.; Lim, D.H.; Jang, K.-T.; Lim, T.; Lee, J.; Choi, Y.-L.; Jang, H.-L.; Yi, J.H.; Baek, K.K.; Park, S.H.; et al. Impact of KRAS mutations on clinical outcomes in pancreatic cancer patients treated with first-line gemcitabine-based chemotherapy. Mol. Cancer Ther. 2011, 10, 1993–1999. [Google Scholar] [CrossRef]
  88. Ogura, T.; Yamao, K.; Hara, K.; Mizuno, N.; Hijioka, S.; Imaoka, H.; Sawaki, A.; Niwa, Y.; Tajika, M.; Kondo, S.; et al. Prognostic value of K-ras mutation status and subtypes in endoscopic ultrasound-guided fine-needle aspiration specimens from patients with unresectable pancreatic cancer. J. Gastroenterol. 2013, 48, 640–646. [Google Scholar] [CrossRef]
  89. Bournet, B.; Muscari, F.; Buscail, C.; Assenat, E.; Barthet, M.; Hammel, P.; Selves, J.; Guimbaud, R.; Cordelier, P.; Buscail, L. KRAS G12D Mutation Subtype Is A Prognostic Factor for Advanced Pancreatic Adenocarcinoma. Clin. Transl. Gastroenterol. 2016, 7, e157. [Google Scholar] [CrossRef]
  90. Qian, Z.R.; Rubinson, D.A.; Nowak, J.A.; Morales-Oyarvide, V.; Dunne, R.F.; Kozak, M.M.; Welch, M.W.; Brais, L.K.; Da Silva, A.; Li, T.; et al. Association of Alterations in Main Driver Genes With Outcomes of Patients With Resected Pancreatic Ductal Adenocarcinoma. JAMA Oncol. 2018, 4, e173420. [Google Scholar] [CrossRef]
  91. Haigis, K.M. KRAS Alleles: The Devil Is in the Detail. Trends Cancer 2017, 3, 686–697. [Google Scholar] [CrossRef]
  92. Ihle, N.T.; Byers, L.A.; Kim, E.S.; Saintigny, P.; Lee, J.J.; Blumenschein, G.R.; Tsao, A.; Liu, S.; Larsen, J.E.; Wang, J.; et al. Effect of KRAS oncogene substitutions on protein behavior: Implications for signaling and clinical outcome. J. Natl. Cancer Inst. 2012, 104, 228–239. [Google Scholar] [CrossRef]
  93. Pantsar, T.; Rissanen, S.; Dauch, D.; Laitinen, T.; Vattulainen, I.; Poso, A. Assessment of mutation probabilities of KRAS G12 missense mutants and their long-timescale dynamics by atomistic molecular simulations and Markov state modeling. PLoS Comput. Biol. 2018, 14, e1006458. [Google Scholar] [CrossRef]
  94. Cristiano, S.; Leal, A.; Phallen, J.; Fiksel, J.; Adleff, V.; Bruhm, D.C.; Jensen, S.Ø.; Medina, J.E.; Hruban, C.; White, J.R.; et al. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature 2019. [Google Scholar] [CrossRef]
  95. Shen, S.Y.; Singhania, R.; Fehringer, G.; Chakravarthy, A.; Roehrl, M.H.A.; Chadwick, D.; Zuzarte, P.C.; Borgida, A.; Wang, T.T.; Li, T.; et al. Sensitive tumour detection and classification using plasma cell-free DNA methylomes. Nature 2018, 563, 579–583. [Google Scholar] [CrossRef]
  96. Barault, L.; Amatu, A.; Siravegna, G.; Ponzetti, A.; Moran, S.; Cassingena, A.; Mussolin, B.; Falcomatà, C.; Binder, A.M.; Cristiano, C.; et al. Discovery of methylated circulating DNA biomarkers for comprehensive non-invasive monitoring of treatment response in metastatic colorectal cancer. Gut 2018, 67, 1995–2005. [Google Scholar] [CrossRef]
  97. Cheon, H.; Paik, J.H.; Choi, M.; Yang, H.-J.; Son, J.-H. Detection and manipulation of methylation in blood cancer DNA using terahertz radiation. Sci. Rep. 2019, 9, 6413. [Google Scholar] [CrossRef]
  98. Couto, N.; Caja, S.; Maia, J.; Strano Moraes, M.C.; Costa-Silva, B. Exosomes as emerging players in cancer biology. Biochimie 2018, 155, 2–10. [Google Scholar] [CrossRef]
  99. Capello, M.; Vykoukal, J.V.; Katayama, H.; Bantis, L.E.; Wang, H.; Kundnani, D.L.; Aguilar-Bonavides, C.; Aguilar, M.; Tripathi, S.C.; Dhillon, D.S.; et al. Exosomes harbor B cell targets in pancreatic adenocarcinoma and exert decoy function against complement-mediated cytotoxicity. Nat. Commun. 2019, 10, 254. [Google Scholar] [CrossRef]
  100. Nazarenko, I.; Rana, S.; Baumann, A.; McAlear, J.; Hellwig, A.; Trendelenburg, M.; Lochnit, G.; Preissner, K.T.; Zöller, M. Cell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. Cancer Res. 2010, 70, 1668–1678. [Google Scholar] [CrossRef]
  101. Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H.; et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 2015, 17, 816–826. [Google Scholar] [CrossRef]
  102. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef]
  103. Becker, A.; Thakur, B.K.; Weiss, J.M.; Kim, H.S.; Peinado, H.; Lyden, D. Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis. Cancer Cell 2016, 30, 836–848. [Google Scholar] [CrossRef] [Green Version]
  104. 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. [Google Scholar] [CrossRef]
  105. Goto, T.; Fujiya, M.; Konishi, H.; Sasajima, J.; Fujibayashi, S.; Hayashi, A.; Utsumi, T.; Sato, H.; Iwama, T.; Ijiri, M.; et al. An elevated expression of serum exosomal microRNA-191, - 21, -451a of pancreatic neoplasm is considered to be efficient diagnostic marker. BMC Cancer 2018, 18, 116. [Google Scholar] [CrossRef]
  106. Madhavan, B.; Yue, S.; Galli, U.; Rana, S.; Gross, W.; Müller, M.; Giese, N.A.; Kalthoff, H.; Becker, T.; Büchler, M.W.; et al. 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. [Google Scholar] [CrossRef]
  107. Melo, S.A.; Luecke, L.B.; Kahlert, C.; Fernandez, A.F.; Gammon, S.T.; Kaye, J.; LeBleu, V.S.; Mittendorf, E.A.; Weitz, J.; Rahbari, N.; et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature 2015, 523, 177–182. [Google Scholar] [CrossRef] [Green Version]
  108. Lai, X.; Wang, M.; McElyea, S.D.; Sherman, S.; House, M.; Korc, M. A microRNA signature in circulating exosomes is superior to exosomal glypican-1 levels for diagnosing pancreatic cancer. Cancer Lett. 2017, 393, 86–93. [Google Scholar] [CrossRef] [Green Version]
  109. Allenson, K.; Castillo, J.; San Lucas, F.A.; Scelo, G.; Kim, D.U.; Bernard, V.; Davis, G.; Kumar, T.; Katz, M.; Overman, M.J.; et al. High prevalence of mutant KRAS in circulating exosome-derived DNA from early-stage pancreatic cancer patients. Ann. Oncol. 2017, 28, 741–747. [Google Scholar] [CrossRef]
  110. Xu, Y.-F.; Hannafon, B.N.; Zhao, Y.D.; Postier, R.G.; Ding, W.-Q. Plasma exosome miR-196a and miR-1246 are potential indicators of localized pancreatic cancer. Oncotarget 2017, 8, 77028–77040. [Google Scholar] [CrossRef]
  111. Yang, K.S.; Im, H.; Hong, S.; Pergolini, I.; Del Castillo, A.F.; Wang, R.; Clardy, S.; Huang, C.-H.; Pille, C.; Ferrone, S.; et al. Multiparametric plasma EV profiling facilitates diagnosis of pancreatic malignancy. Sci. Transl. Med. 2017, 9, eaal3226. [Google Scholar] [CrossRef] [Green Version]
  112. Lewis, J.M.; Vyas, A.D.; Qiu, Y.; Messer, K.S.; White, R.; Heller, M.J. Integrated Analysis of Exosomal Protein Biomarkers on Alternating Current Electrokinetic Chips Enables Rapid Detection of Pancreatic Cancer in Patient Blood. ACS Nano 2018, 12, 3311–3320. [Google Scholar] [CrossRef]
  113. Jin, H.; Liu, P.; Wu, Y.; Meng, X.; Wu, M.; Han, J.; Tan, X. Exosomal zinc transporter ZIP4 promotes cancer growth and is a novel diagnostic biomarker for pancreatic cancer. Cancer Sci. 2018, 109, 2946–2956. [Google Scholar] [CrossRef] [Green Version]
  114. European Study Group on Cystic Tumours of the Pancreas. European evidence-based guidelines on pancreatic cystic neoplasms. Gut 2018, 67, 789–804. [Google Scholar] [CrossRef]
  115. Karasek, P.; Gablo, N.; Hlavsa, J.; Kiss, I.; Vychytilova-Faltejskova, P.; Hermanova, M.; Kala, Z.; Slaby, O.; Prochazka, V. Pre-operative Plasma miR-21-5p Is a Sensitive Biomarker and Independent Prognostic Factor in Patients with Pancreatic Ductal Adenocarcinoma Undergoing Surgical Resection. Cancer Genom. Proteom. 2018, 15, 321–327. [Google Scholar] [CrossRef] [Green Version]
  116. Humeau, M.; Vignolle-Vidoni, A.; Sicard, F.; Martins, F.; Bournet, B.; Buscail, L.; Torrisani, J.; Cordelier, P. Salivary MicroRNA in Pancreatic Cancer Patients. PLoS ONE 2015, 10, e0130996. [Google Scholar] [CrossRef]
  117. Buscail, L.; Bournet, B.; Vernejoul, F.; Cambois, G.; Lulka, H.; Hanoun, N.; Dufresne, M.; Meulle, A.; Vignolle-Vidoni, A.; Ligat, L.; et al. First-in-man Phase 1 Clinical Trial of Gene Therapy for Advanced Pancreatic Cancer: Safety, Biodistribution, and Preliminary Clinical Findings. Mol. Ther. 2015, 23, 779–789. [Google Scholar] [CrossRef] [Green Version]
  118. Cacheux, J.; Brut, M.; Bancaud, A.; Cordelier, P.; Leïchlé, T. Spatial Analysis of Nanofluidic-Embedded Biosensors for Wash-Free Single-Nucleotide Difference Discrimination. ACS Sens. 2018, 3, 606–611. [Google Scholar] [CrossRef]
  119. Thakur, B.K.; Zhang, H.; Becker, A.; Matei, I.; Huang, Y.; Costa-Silva, B.; Zheng, Y.; Hoshino, A.; Brazier, H.; Xiang, J.; et al. Double-stranded DNA in exosomes: A novel biomarker in cancer detection. Cell Res. 2014, 24, 766–769. [Google Scholar] [CrossRef]
  120. Kahlert, C.; Melo, S.A.; Protopopov, A.; Tang, J.; Seth, S.; Koch, M.; Zhang, J.; Weitz, J.; Chin, L.; Futreal, A.; et al. Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J. Biol. Chem. 2014, 289, 3869–3875. [Google Scholar] [CrossRef]
  121. Le Calvez-Kelm, F.; Foll, M.; Wozniak, M.B.; Delhomme, T.M.; Durand, G.; Chopard, P.; Pertesi, M.; Fabianova, E.; Adamcakova, Z.; Holcatova, I.; et al. KRAS mutations in blood circulating cell-free DNA: A pancreatic cancer case-control. Oncotarget 2016, 7, 78827–78840. [Google Scholar] [CrossRef]
  122. Lu, H.; Niu, F.; Liu, F.; Gao, J.; Sun, Y.; Zhao, X. Elevated glypican-1 expression is associated with an unfavorable prognosis in pancreatic ductal adenocarcinoma. Cancer Med. 2017, 6, 1181–1191. [Google Scholar] [CrossRef]
  123. Kalluri, R. The biology and function of exosomes in cancer. J. Clin. Investig. 2016, 126, 1208–1215. [Google Scholar] [CrossRef]
  124. Qian, L.; Yu, S.; Chen, Z.; Meng, Z.; Huang, S.; Wang, P. Functions and clinical implications of exosomes in pancreatic cancer. Biochim. Biophys. Acta Rev. Cancer 2018, 1871, 75–84. [Google Scholar] [CrossRef]
  125. Sol, N.; Wurdinger, T. Platelet RNA signatures for the detection of cancer. Cancer Metastasis Rev. 2017, 36, 263–272. [Google Scholar] [CrossRef]
  126. Best, M.G.; Sol, N.; Kooi, I.; Tannous, J.; Westerman, B.A.; Rustenburg, F.; Schellen, P.; Verschueren, H.; Post, E.; Koster, J.; et al. RNA-Seq of Tumor-Educated Platelets Enables Blood-Based Pan-Cancer, Multiclass, and Molecular Pathway Cancer Diagnostics. Cancer Cell 2015, 28, 666–676. [Google Scholar] [CrossRef] [Green Version]
  127. Kuznetsov, H.S.; Marsh, T.; Markens, B.A.; Castaño, Z.; Greene-Colozzi, A.; Hay, S.A.; Brown, V.E.; Richardson, A.L.; Signoretti, S.; Battinelli, E.M.; et al. Identification of luminal breast cancers that establish a tumor-supportive macroenvironment defined by proangiogenic platelets and bone marrow-derived cells. Cancer Discov. 2012, 2, 1150–1165. [Google Scholar] [CrossRef]
  128. Kim, J.; Bamlet, W.R.; Oberg, A.L.; Chaffee, K.G.; Donahue, G.; Cao, X.-J.; Chari, S.; Garcia, B.A.; Petersen, G.M.; Zaret, K.S. Detection of early pancreatic ductal adenocarcinoma with thrombospondin-2 and CA19-9 blood markers. Sci. Transl. Med. 2017, 9, eaah5583. [Google Scholar] [CrossRef]
  129. Berger, A.W.; Schwerdel, D.; Reinacher-Schick, A.; Uhl, W.; Algül, H.; Friess, H.; Janssen, K.-P.; König, A.; Ghadimi, M.; Gallmeier, E.; et al. A Blood-Based Multi Marker Assay Supports the Differential Diagnosis of Early-Stage Pancreatic Cancer. Theranostics 2019, 9, 1280–1287. [Google Scholar] [CrossRef]
  130. Capello, M.; Bantis, L.E.; Scelo, G.; Zhao, Y.; Li, P.; Dhillon, D.S.; Patel, N.J.; Kundnani, D.L.; Wang, H.; Abbruzzese, J.L.; et al. Sequential Validation of Blood-Based Protein Biomarker Candidates for Early-Stage Pancreatic Cancer. J. Natl. Cancer Inst. 2017, 109. [Google Scholar] [CrossRef]
  131. Fahrmann, J.F.; Bantis, L.E.; Capello, M.; Scelo, G.; Dennison, J.B.; Patel, N.; Murage, E.; Vykoukal, J.; Kundnani, D.L.; Foretova, L.; et al. A Plasma-Derived Protein-Metabolite Multiplexed Panel for Early-Stage Pancreatic Cancer. J. Natl. Cancer Inst. 2019, 111, 372–379. [Google Scholar] [CrossRef]
  132. Mayerle, J.; Kalthoff, H.; Reszka, R.; Kamlage, B.; Peter, E.; Schniewind, B.; González Maldonado, S.; Pilarsky, C.; Heidecke, C.-D.; Schatz, P.; et al. Metabolic biomarker signature to differentiate pancreatic ductal adenocarcinoma from chronic pancreatitis. Gut 2018, 67, 128–137. [Google Scholar] [CrossRef]
  133. Bian, B.; Fanale, D.; Dusetti, N.; Roque, J.; Pastor, S.; Chretien, A.-S.; Incorvaia, L.; Russo, A.; Olive, D.; Iovanna, J. Prognostic significance of circulating PD-1, PD-L1, pan-BTN3As, BTN3A1 and BTLA in patients with pancreatic adenocarcinoma. Oncoimmunology 2019, 8, e1561120. [Google Scholar] [CrossRef]
Figure 1. Clinical validity of circulating tumour elements in pancreatic cancer patients as reported in published major clinical studies. Yellow circles: absolute numbers of publications; Blue circles: number of publications with a significant correlation between diagnosis and the presence of circulating tumour elements; orange circles: publications with a significant correlation between response to neoadjuvant therapy and the presence of circulating tumour elements; light red circles: publications with a significant correlation between prognosis and the presence of circulating tumour elements. CTCs: circulating tumour cells; ctDNA: circulating tumour DNA; EVs: extracellular vesicles.
Figure 1. Clinical validity of circulating tumour elements in pancreatic cancer patients as reported in published major clinical studies. Yellow circles: absolute numbers of publications; Blue circles: number of publications with a significant correlation between diagnosis and the presence of circulating tumour elements; orange circles: publications with a significant correlation between response to neoadjuvant therapy and the presence of circulating tumour elements; light red circles: publications with a significant correlation between prognosis and the presence of circulating tumour elements. CTCs: circulating tumour cells; ctDNA: circulating tumour DNA; EVs: extracellular vesicles.
Cancers 11 00852 g001
Table 1. Results of main clinical pilot studies assessing performances of circulating tumour cells (CTCs) detection in the diagnosis and prognosis of pancreatic ductal adenocarcinoma (PDAC).
Table 1. Results of main clinical pilot studies assessing performances of circulating tumour cells (CTCs) detection in the diagnosis and prognosis of pancreatic ductal adenocarcinoma (PDAC).
PDAC Patient Number (Control)Type of Tumour: Resected, Locally Advanced, Metastatic, AllCTC EnrichmentCTC DetectionCTC CountCTC Detection Rate in PDAC PatientsPrognosis Value of CTCsReference
20 (15 benign diseases)All (Samples before treatment)Density centrifugationRT-PCR CEANA26%Positive correlation with recurrenceMataki et al., 2004
[29]
154 (68 benign diseases)All (Samples before treatment)Density centrifugationRT-PCR CK20NA34%Shorter OS (meta.) (p = 0.05)Soeth et al., 2005
[30]
25 (15 benign diseases)All (Samples before treatment)Immunomagnetic (EpCAM)RT-PCR: cMET, hTERT, CK20, CEANA80–100% (sensitivity 100%; specificity 96%)Not studiedZhou et al., 2009
[32]
41 (20 HC)All (Sample before and post treatment)Immunomagnetic (leukocytes CD45+ depletion)ICC: CK8/CK18+, CA19-9+, CD451680% before and 20% after chemotherapyNot studiedRen et al., 2011
[33]
48 (10 CP)All (Samples before and after treatment)NoneReal-time RT-PCR mRNA EpCAMNA25% pre-operative
65% post-operative
No correlation with any outcomeSergeant et al., 2011
[31]
54 (No)All (Sample time: NA)Immunomagnetic: ISET and CellSearch®ISET: Cytology, CD45
ICC: CK+, DAPI+, CD45
- ISET: 26
- CellSearch®:6
ISET:93% CellSearch®:40%No correlation with any outcomeKhoja et al., 2012
[34]
79 (No)LA (Samples before and after chemotherapy)Immunomagnetic: CellSearch®ICC: CK+, DAPI+, CD451 to 15 (only 1 or 2 patients)11%Poor differentiation and shorter OS (p = 0.01)Bidard et al., 2013
[52]
72 (28 benign diseases)All (Samples before treatment)Microfluidic (NanoVelcro)ICC: CK+, DAPI+, CD45 KRAS mutation0 to ≥5 (*)75%≥3 CTCs: discriminate metastatic disease (p < 0.001)Ankeny et al., 2016
[35]
48 (No)Metastatic (Samples before treatment)Immunomagnetic: CellSearch®ICC: CK+, DAPI+, CD45, MUC-1+23 patients: ≥1
9 patients: ≥2
48%CTC MUC-1+ correlate with a shorter OS (p = 0.044)Dotan et al., 2016
[36]
60 (no)All (40% of the samples performed after neo-adjuvant therapy)Size based ISETICC: CK+, ALDH+, CD133+, CD44+Mean: 7.1
Median: 4
78%CK+/ALDH+: shorter OS and DFS CK+/CD133+/CD 44+: shorter DFSPoruk et al., 2017
[37]
58 PDAC (10 HC)All (samples time NA)Size based: Screencell©Cytology KRAS mutationRange 0–1367%>3 CTC+: shorter OSKuleman et al., 2017
[38]
52 (10 benign diseases)All (samples time NA)Size based Screencell©CytologyMedian 4
Range 0–151
67%No correlationSefrioui et al., 2017
[43]
65 (15 HC)LA and Meta. (Samples before treatment)Immunomagnetic CellSearch®ICC: CK+, DAPI+, CD454.932.3%Independent predictor of shorter OSOkubo et al., 2017
[39]
100 (26 benign diseases)All (32% of the samples after neo-adjuvant therapy)Microfluidic Nano-velcroICC: CK+, DAPI+, CD45NA78%Correlated with presence of occult metastasis, shorter PFS and OSCourt et al., 2018
[40]
69 (9 benign diseases)All (10% of the samples after neo-adjuvant therapy)Immunomagnetic MACS and CellSearch® (n = 20)ICC: CK+, DAPI+, CD4517 patients >1
13 patients >2
33.3%Independent predictor of shorter PFS and OSEffenberger et al., 2018
[41]
242 (No)All (sample time NA)Immunomagnetic CellSearch®ICC: CK+, DAPI+, CD45Median 1
Range 1–33
78.5%Shorter PFS (p < 0.001)Hugenschmidt et al., 2018
[42]
24 (no)Metastatic (Samples before and after chemotherapy)MicrofluidicICC: CK+, DAPI+, CD133, EpCAM+, CD45-Mean 3.87 CTCs/mL84.4%No correlationVarillas et al., 2019
[44]
100 (16 benign disease, 30 HC)All (Samples before and after treatment)MicrofluidicICC: Vimentin+, EpCAM+, CD45-Median 3
Range 0–23
76%≥2 CTCs vimentin+: correlate with a shorter PFSWei et al., 2019
[45]
CTCs: circulating tumour cells; LA: locally advanced PDAC; Metastatic: metastatic PDAC: All: resected + locally advanced + metastatic PDAC patients; NA: not available; HC: healthy control; CP: chronic pancreatitis; MACS: magnetic activation cell search; ISET: isolation by size of epithelial tumour cells; ICC: immuno-cyto-chemistry; DAPI: 4’, 6-diamidino-2-phénylindole as fluorescent protein linking to thymine and adenine DNA bases; OS: overall survival rate; PFS: progression free survival; DFS: disease free survival; NA: not available; MUC: mucin; EpCAM: epithelial cell adhesion molecule; ALDH: aldehyde dehydrogenase. CTC count is mostly expressed as mean/7.5 mL. (*): 18 patients: 0 CTC; 54 patients: ≥1 CTC; 39 patients: ≥2 CTCs; 29 patients: ≥3 CTCs; 18 patients: ≥5 CTCs.
Table 2. The main studies that have investigated the role of ctDNA in the diagnosis and/or prognosis of pancreatic ductal carcinoma.
Table 2. The main studies that have investigated the role of ctDNA in the diagnosis and/or prognosis of pancreatic ductal carcinoma.
PDAC Patient Number (Control)Type of Tumour: Resected, Locally Advanced, Metastatic, AllSiteTarget for ctDNA% of Mutations or Genetic Alterations in PDAC PatientsDiagnosis PerformancesPositive Correlation with a Poor Prognosis (OS) (p) *Reference
44 (60: 37 CP and 23 miscellaneous)AllPlasmaKRAS mutation
Amplified PCR
27Sensitivity: 27%
Specificity: 100%
Yes—<0.005Castells et al., 1999
[55]
47 (31: CP)AllSerumKRAS mutation sequencing47Sensitivity: 47%
Specificity: 87%
No—NsMaire et al., 2002
[56]
56 (13: CP)AllPlasmaKRAS mutation
PNA-mediated PCR
clamping and real-time PCR
36Sensitivity: 36%
Specificity: 100%
No—0.10Däbritz et al., 2009
[57]
91 (No)LA + Meta.PlasmaKRAS mutation sequencing33-Yes—<0.001Chen et al., 2010
[58]
36 (49: 25 HC and 24 miscellaneous)AllPlasmaKRAS mutation cold-PCR combined with an unlabelled-probe HRM72Sensitivity: 81%
Specificity: 87.5%
-Wu et al., 2014
[59]
27 (No)LA + Meta.PlasmaKRAS mutation
ARMS PCR
37-Yes—0.003
Yes—0.014 ***
Semrad et al., 2015
[60]
51 (No)RPlasmaKRAS mutation
dPCR
43Sensitivity: 43%
Specificity: >99%
Yes (predictor of disease recurrence)—0.015Sausen et al., 2015
[61]
45 (No)AllPlasmaKRAS mutation
dPCR
26-Yes—0.001Earl et al., 2015
[62]
110 (25: HC)AllPlasmaKRAS mutation
RFLP + sequ.
Two-step enriched-nested PCR
31-No—0.36Singh et al., 2015
[63]
75 (40: 20 CP and 20 HC)AllSerumKRAS mutation
dPCR
63-Yes—0.024Kinugasa et al., 2015
[64]
259 (No)AllPlasmaKRAS mutation
dPCR
8 (R), 18 (LA), 59 (M)-Yes—<0.0001Takai et al., 2015
[65]
105 (20 HC)RPlasmaKRAS mutation
dPCR
31-Yes—<0.0001
Yes—<0.001 **
Hadano et al., 2016
[71]
40 (10 HC)AllPlasma and serumKRAS mutation
dPCR
48 (All)
38 (LA)
63 (LA and Meta. Serum)
-Yes—<0.01Ako et al., 2016
[67]
188 (No)MetPlasmaKRAS mutation
dPCR
83-Yes—0.019Cheng et al., 2017
[68]
135 (No)AllPlasmaKRAS mutation
NGS/dPCR
41 (LA and Meta.)-LA + Met: Yes—p < 0.001
Resected: Yes—0.027; Yes—0.03 **
Pietrasz et al., 2017
[69]
60 (No)LA + Meta.PlasmaKRAS mutation BEAMing65-Yes—0.001
Yes—0.0022 **
Van Laethem et al., 2017
[70]
95 (No)AllPlasma28 genes
Methylation-specific PCR
27 (>10 hypermethylated genes)-Yes Henriksen et al., 2017
[85]
26 (26: 14 CP and 12 HC)AllPlasmaKRAS mutation dPCR
NGS: KRAS, SMAD4, CDKN2A and TP53
NGS: 27
dPCR: 23
-Yes—0.018 ****Adamoet al., 2017
[66]
27 (43 HC)LA + Meta.PlasmaKRAS mutation
dPCR
70.4-No—0.16—0.24 ***Del Re et al., 2017
[72]
221 (182 HC)RPlasmaKRAS mutation
PCR Safe-Sequencing System
30Sensitivity: 30%
Specificity: 99.5%
-Cohen et al., 2017
[73]
34 (No)AllPlasmaNGS: 25 genes (including KRAS)25 genes: 74
KRAS only: 29
-Yes—0.045Pishvaian et al., 2017
[74]
106 (No)AllPlasmaKRAS mutation dPCR68(R), 72(LA), 87(M)Sensitivity: 78%
Specificity: 33%
Yes—0.008
Yes—0.003 ***
Kim et al., 2018
[75]
65 (20 HC)AllPlasmaKRAS mutation
dPCR
80-No—0.73Lin et al., 2018
[77]
45 (No)RSerumKRAS mutation teal-time quantitative PCR55-Pre-operative samples: No—0.258–0.710 **
Post-operative samples: Yes—0.027 **
Nakano et al., 2018
[76]
* A worse prognosis in patients with a mutated KRAS vs. wild type in term of overall survival (OS). **: disease-free survival. ***: progression-free survival. ****: disease specific survival. CP: chronic pancreatitis; HC: healthy controls; R: resected PDAC; LA: locally advanced PDAC; M or Meta.: metastatic PDAC: All: resected + locally advanced + metastatic PDAC patients; RFLP: restriction fragment length polymorphism; dPCR: digital droplet PCR; NGS: next generation sequencing; BEAM: Beads Emulsion Amplification Magnetic; ARMS PCR: amplification-refractory mutation system polymerase chain reaction; Cold-PCR: co-amplification at lower denaturation temperature-PCR; PNA-mediated PCR: peptide nucleic acid-mediated PCR; HRM: high resolution melt.
Table 3. Results of main clinical pilot studies assessing performances of exosome detection in PDAC.
Table 3. Results of main clinical pilot studies assessing performances of exosome detection in PDAC.
Patient Number (PDAC)Type of Tumour: Resected, Locally Advanced, Metastatic, All (Treatment)Molecular Target(s)Method of IsolationExosomes Detection Rate in PDAC PatientsExosomes Diagnosis PerformancesExosomes Prognosis ValueReference
16
(6 HP, 6 CP, 5 cysts, 5 ampullary carcinoma)
All (12 metastatic)miR-17-5p, -21, -155Ultracentrifugation
RT qPCR
NA(**)miR-17-5p correlated with metastasisQue et al., 2013
[104]
131
(64 HC)
AllmiR-1246, -4644, -3976, -4306; CD44v6, TSPAN8, EpCAM, MET, CD104Sucrose gradient, micro-array, RTqPCR, flow cytometry, latex beadsNASens. 100%
Spec. 80%
NSMadhavan et al., 2014
[106]
146
(benign pancreatic diseases 32, 120 HC)
All (Neo-adjuvant: 10)GPC1Latex beads
Ultracentrifugation
Ac GPC1
100%Sens. 100%
Spec. 100%
GPC1+ correlates with worse DFS and OSMelo et al., 2015
[107]
29
(CP 11)
Resected and locally advancedGPC1 miR-10b, -21, -30c, -181a, -let7aGPC1 LC-MS/ML
RT qPCR
100%Sens. 100%
Spec. 100%
NSLai et al., 2017
[108]
127
(136 HC)
AllExo DNA ctDNAUltracentrifugation
Flowcytometry
dPCR
54%Sens. 54%
Spec. 84%
PPV 76%
NPV 66%
Worse DFS
P = 0.03
RR: 4.68
441 days vs. 127
Allenson et al., 2017
[109]
15
(15 HC)
AllmiR: R196a, 196b and 1246ExoKit
RTqPCR
NGS
Significantly higher for 196a and 1246AUC:
196a: 0.81
1246: 0.73
196b 0.71
NSXu et al., 2017
[110]
68
(41 benign pancreatic diseases;18 HC)
All (Neo-adjuvant: 33)Signature: EGRF, EpCAM, MUC1, GPC1, WNT2Ultracentrifugation89%Sens. 86%
Spec. 81%
NSYang et al., 2018
[111]
20
(20 benign diseases)
Resected and locally advancedProtein CD63, GPC1AC electrokinetics
immunofluorescence
Significantly higher in PDAC cohortSens. 99
Spec. 82
NSLewis et al., 2018
[112]
32
(IPMN 29, 22 HC)
AllmiR-191, -21, -451aExoKit Quick
NGS RT qPCR
(*)miR21 worse OSGoto et al., 2018
[105]
24
(14 CP, 50 miscellaneous, 46 HC)
NAProtein ZIP4Exo Kit precipitationSignificantly higher in PDACAUC ROC curve 0.89NSJin et al., 2018
[113]
194
(25 cysts, 12 HC)
All (123 metastatic)Exo DNA KRASUltracentrifugation ddPCR61% metastatic
38% resectable
NSMAF >5%
Predictor PFS OS
Bernard et al., 2019
[79]
miR: microRNA; CP: chronic pancreatitis; HC: healthy control patients; IPMN: intraductal papillary mucinous neoplasia; NET: neuroendocrine tumour; NA: not available; NS: not studied; NGS: next generation sequencing; GPC1: sulfate proteoglycan 1; AUC: area under ROC curves; EpCAM; epithelial cell adhesion molecule; MUC1: mucin 1; TSPAN8: tetraspanin8; MAF: mutation allelic frequency. (*): miR-191: Sens. 71.9%, Spec. 84.2%, accuracy 76.6%; miR-21: Sens. 80.7%, Spec. 81%, accuracy 80.8%; miR-451a: Sens. 65.8%, Spec. 85.7%, accuracy 73.6%. (**): miR-17-5p: Sens. 72.7%, Spec. 92.6%; miR-21: Sens 95.5%, Spec. 81.5%.

Share and Cite

MDPI and ACS Style

Buscail, E.; Maulat, C.; Muscari, F.; Chiche, L.; Cordelier, P.; Dabernat, S.; Alix-Panabières, C.; Buscail, L. Liquid Biopsy Approach for Pancreatic Ductal Adenocarcinoma. Cancers 2019, 11, 852. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers11060852

AMA Style

Buscail E, Maulat C, Muscari F, Chiche L, Cordelier P, Dabernat S, Alix-Panabières C, Buscail L. Liquid Biopsy Approach for Pancreatic Ductal Adenocarcinoma. Cancers. 2019; 11(6):852. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers11060852

Chicago/Turabian Style

Buscail, Etienne, Charlotte Maulat, Fabrice Muscari, Laurence Chiche, Pierre Cordelier, Sandrine Dabernat, Catherine Alix-Panabières, and Louis Buscail. 2019. "Liquid Biopsy Approach for Pancreatic Ductal Adenocarcinoma" Cancers 11, no. 6: 852. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers11060852

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

Article Metrics

Back to TopTop