Liquid biopsy in pancreatic cancer: the beginning of a new era.
Journal: 2018/November - Oncotarget
ISSN: 1949-2553
Abstract:
With dismal survival rate pancreatic cancer remains one of the most aggressive and devastating malignancy. Predominantly, due to the absence of a dependable methodology for early identification and limited therapeutic options for advanced disease. However, it takes over 17 years to develop pancreatic cancer from initiation of mutation to metastatic cancer; therefore, if diagnosed early; it may increase overall survival dramatically, thus, providing a window of opportunity for early detection. Recently, genomic expression analysis defined 4 subtypes of pancreatic cancer based on mutated genes. Hence, we need simple and standard, minimally invasive test that can monitor those altered genes or their associated pathways in time for the success of precision medicine, and liquid biopsy seems to be one answer to all these questions. Again, liquid biopsy has an ability to pair with genomic tests. Additionally, liquid biopsy based development of circulating tumor cells derived xenografts, 3D organoids system, real-time monitoring of genetic mutations by circulating tumor DNA and exosome as the targeted drug delivery vehicle holds lots of potential for the treatment and cure of pancreatic cancer. At present, diagnosis of pancreatic cancer is frantically done on the premise of CA19-9 and radiological features only, which doesn't give a picture of genetic mutations and epigenetic alteration involved. In this manner, the current diagnostic paradigm for pancreatic cancer diagnosis experiences low diagnostic accuracy. This review article discusses the current state of liquid biopsy in pancreatic cancer as diagnostic and therapeutic tools and future perspectives of research in the light of circulating tumor cells, circulating tumor DNA and exosomes.
Relations:
Content
Citations
(11)
Similar articles
Articles by the same authors
Discussion board
Oncotarget. May/31/2018; 9(42): 26900-26933
Published online May/31/2018

Liquid biopsy in pancreatic cancer: the beginning of a new era

Abstract

INTRODUCTION

Pancreatic cancer (PC) remains one of the most deadly malignancies with an overall five-year survival probability less than 7% in all stages combined [1]. Moreover, it is the fourth leading cause of cancer-related death in the United States, with 53,670 new cases and estimated 43,090 deaths in 2017 [2]. In China, the estimated incidence and death of the PC is 90,100 and 79,400, respectively [3]. Surgical resection is only the main curative treatment; in any case, because of late-presenting clinical features, roughly 30 to 40 percent have the locally advanced disease and another 40 percent have a metastatic tumor at the time of diagnosis. Thus, palliative chemotherapy remains the main treatment option for most of these patients [46].

Recently, advances in understanding of the molecular pathology of the PC have given hope to new therapeutic approaches; however, according to recently published systematic review, lack of clinical meaningful trials in the past 25 years might be a reason behind the failure to achieve improvements in early diagnosis, management, and prolongation of overall survival of the PC [7].

With growing research, it is now well understood that the PC is a genetic disease, with complex mutation of cancer genes, and the progression of cancer is characterized by high heterogeneity [810]. In earlier studies, KRAS, TP53, CDKN2A, and SMAD4 have been identified as recurrently mutated genes in PC [11, 12]. These discoveries have enhanced our understanding of the molecular pathology of the PC. Recently, genomic analyses have identified different molecular subtypes of PC based on the expression of transcriptional profiles and the structural variations [8, 1315]. Thus, it's very crucial for early detection of mutant genes to identify PC and its subtypes, for effective management strategy of the disease. In the past decades, numerous studies have shown the potential clinical utility of liquid biopsy, such as circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and circulating tumor exosomes for various cancers, including PC [1621]. These promising markers serve as a unique approach for early detection, monitoring and managing disease states. In recent years, noninvasive disease monitoring technology has witnessed an extraordinary explosion of research in the field of liquid biopsy since circulating cell free DNA (cfDNA) was first revealed in body fluids by Mandel and Metais in 1948 [22]. In this review, we have outlined better understanding of different components of liquid biopsy, especially CTCs, ctDNA and exosomes and their potential clinical utility for PC patients. Moreover, we have also drafted numerous of the rational challenges come across using the liquid biopsy techniques.

CURRENT STATUS IN DIAGNOSIS OF PC

Recently, studies have suggested that pancreatic cancer takes over 17 years to develop, from initiation of mutation in the gene to metastatic cancer, trailed by death roughly after 2.7 years [6, 23, 24]; therefore, if diagnosed early; it may increase overall survival dramatically, and thus, provide a window of opportunity for early detection. Currently, there is no official PC screening program, a confinement of screening for early PC is the absence of sensitive and specific markers [25]. Most commonly used blood-based tumor biomarkers in clinical practice are carbohydrate antigen (CA) 19-9 and carcinoembryonic antigen (CEA). Besides, CA19-9 is the only one currently recommended for clinical use by the NCCN guidelines for PC [26]. According to a recent meta-analysis, CA 19-9 has satisfying pooled specificity while the poor pooled sensitivity for differentiating benign from malignant pancreatic tumors, the pooled sensitivity and specificity were 0.47 (95% CI: 0.35–0.59), and 0.88 (95% CI: 0.86–0.91), respectively [27]. Additionally, it is not tumor specific and is elevated in many hepatobiliary cancers likewise in benign biliary obstruction [28]. In spite of advances in the molecular pathology of the PC, there is no dependable biomarker, the sensitivity and specificity of these currently used tumor biomarkers are definitely not adequate for the early recognition of PC [29, 30].

At present the diagnosis and staging of the PC to a great extent depends on imaging modalities, including ultrasonography (USG), computed tomography (CT), endoscopic retrograde cholangiopancreatography (ERCP), positron emission tomography (PET), magnetic resonance imaging (MRI), magnetic resonance cholangiopancreatography (MRCP), and endoscopic ultrasonography (EUS) [3137]. In any case, little metastases are hard to detect regardless of the possibility that blends of these modalities are utilized. What's more, these modalities require costly equipment and specialists which are more challenging [35].

Additionally, histological diagnosis often requires invasive tests before surgery. Moreover, because of the difficult anatomical position of the pancreas, a biopsy is often guided by EUS. To the date, endoscopic ultrasonography with fineneedle aspiration (EUSFNA) remains the gold standard in the workup of patients with PC for obtaining the biopsy, with the pooled sensitivity and specificity of 86.8% and 95.8%, respectively [38]. However, EUS-FNA requires sedation and is associated with the number of complications such as tumor seeding along the biopsy tract, pancreatitis, hemorrhage, bowel perforation and aspiration [3942]. In addition, due to the dense desmoplastic reaction in a PC, the majority of the tumor mass is made up of stromal cells instead of the epithelial cancer cells. Thus, giving rise to false negative results, necessitating frequent repetitive biopsies [1, 41, 43]. Hence, the diagnosis is compellingly done on the premise of CA19-9 and radiological features only. In this manner, the current diagnostic paradigm for the diagnosis of PC experiences low diagnostic accuracy.

Consequently, it is urgent to develop new and improved strategies which can address all the above obstacles and identify primary tumors at an early and resectable stage with greater diagnostic sensitivity in vitro, whereas patients with advanced disease must be preoperatively analyzed to dodge surgical impairments and to choose appropriate treatments to enhance the nature of residual life.

MOLECULAR PATHOLOGY OF PC

Genomic analyses of cancer show that there is a complex mutational landscape and genetic stability of cancer cells are compromised, and PC is no exception to this [8, 24]. Additionally, various genetic modifications occur during the development of the PC, including an increase in duplicate chromosomal number, genetic diversification, amplifications and homozygous deletions, recapitulation of clonal expansion, clonal selection, a small subset of driver mutations and loss of heterozygosity with or without duplicate number reduction [6, 24, 4451]. Besides, KRAS, p16, SMAD4, CDKN2A, and TP53, are most commonly mutated genes in the majority of the PC patients, particular KRAS mutations occur in almost 92% of the PC cases [8, 11, 12, 52]. In recent years, genomic analyses have an emphasis on the recognition of somatic mutations and other genetic alterations to identify different molecular subtypes of PC. As a breakthrough, some recent studies have defined subtypes of PC based on the expression of transcriptional profiles and the structural variations [8, 1315].

Collisson et al. [13] classified 3 subtypes of PC 1. Classical- Increased expression of adhesion-associated and epithelial genes, e.g. transmembrane protein 45B (TMEM45B), trefoil factor 1 (TFF1) and mucin 13 (MUC13) 2. Quasi-mesenchymal- Increased expression of mesenchyme-associated genes, e.g. Absent in melanoma 2 (AIM2), glycoprotein m6b (GPM6B) and 5’-nucleotidase ecto (NT5E), and 3. Exocrine like- Increased expression of tumor cell-derived digestive enzyme genes, e.g. Islet-derived 1 beta (REG1B), pancreatic lipase-related protein 2 (PNLIPRP2), and cystic fibrosis transmembrane conductance regulator (CFTR).

Likewise, Moffitt et al. [14] proposed 4 subtypes of the PC: 1. Normal stroma (high expression of ACTA2, VIM, and DES) 2. Activated stroma (high expression of ITGAM, CCL13, CCL18, SPARC, WNT2, WNT5A, MMP9, and MMP11) 3. Classical (high expression of genes such as BTNL8, FAM3D, and ATAD4), and 4. Basal-like (activation of genes such as VGLL1, UCA1, and S100A2). However, basal-like and classical subtypes were also seen in both the normal stroma and activated stroma subtypes.

In addition, Waddell et al. [15] classified 4 subtypes PC based on the structural variation in the mutational landscape 1. Stable (less than 50 structural variations) 2. Locally rearranged (at the minimum of 50 somatic events in the tumor) 3. Scattered (50 to 200 structural variations), and 4. Unstable (more than 200 structural variations).

More recently, genomic expression analysis by Bailey et al. also defined 4 subtypes of the PC: 1. Squamous (TP53 and KDM6A) 2. Pancreatic progenitor (FOXA2/3, PDX1, and MNX1) 3. Immunogenic (upregulated immune networks), and 4. Aberrantly differentiated endocrine exocrine (KRAS, NR5A2, RBPJL, NEUROD1, and NKX2-2) [8].

Nonetheless, the interpretation of this molecular subtyping into the clinical setup has been questioned by conflicting outcomes between these studies. Therapeutic agents that can target these subtypes of the PC and their altered genes or their associated pathways may assume a crucial part in the success of precision medicine for the treatment of the PC [5356].

LIQUID BIOPSY AS A GAME CHANGER

Recently, analysts at The University of Texas MD Anderson Cancer Center have indicated that PC is ready for investigation with a liquid biopsy [57]. A liquid biopsy is simple and painless, minimally invasive sampling and analysis of liquid biomarkers that can be isolated from body fluids, primarily blood [19, 58]. Moreover, liquid biopsies have turned out to be all the more clinically valuable in recent years due to the ability to pair tests on circulating tumor cells (CTCs), circulating tumor nucleic acids (ctNAs) and tumor-derived exosomes with genomic tests [5861] (Figure 1A). Interestingly, a liquid biopsy can characterize tumor biomarkers, similar to tissue biopsy, which allows early detection of disease, real-time evaluation of metastasis, treatment monitoring, empowers examination of primary tumors, and metastases. Additionally, it enables evaluation of tumor, heterogeneity, cancer dormancy, and monitoring of tumor progression along with prognosis [58, 60, 6266]. Recently, liquid biopsy that identifies epidermal growth factor receptor (EGFR) gene mutations in non-small cell lung cancers have been approved by FDA [67, 68].

Figure 1

Application of circulating biomarkers

(A) Application of blood-based liquid biopsy analysis over the span of pancreatic cancer management, peripheral venous blood is collected from the patients for isolation of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA) and exosomes. These circulating biomarkers may be applied to guide initial diagnosis, treatment monitoring or planning, prognosis prediction and developing a new targeted therapy for patients with pancreatic cancer. (B) Functional studies with CTCs and development of CTC— derived xeno-grafts (CDXs), patient-derived tumor xeno-graft (PDTX) and 3D organoids model from CTCs for dynamic monitoring of PC and development of new targeted drugs after its molecular characterization and genomic analysis. (C) Clinical application of ctDNA as a tool for therapy monitoring. ctDNA can be obtained from plasma for genomic analysis, drug testing and use in personalized medicine according to the genomic and epigenomic alteration. (D) Clinical use of exosomes for drug development after genomic and immunological testing. Moreover, use of exosome as a drug delivery vehicle where it can be loaded with drugs, siRNAs, gene etc.

CIRCULATING TUMOR CELLS (CTCS)

Circulating tumor cells (CTCs), represent tumor cells that contain a heterogeneous population of cells, including apoptotic tumor and viable tumor cells that have cast off into the circulation or lymphatic vessels from a primary or metastatic tumor and are transported around the body by undergoing phenotypic changes that are accompanied by a process called as epithelial-mesenchymal transition (EMT) [6973]. Evidence now suggests that the tumors have ability to make their own blood vessels when they reach the size of 1–2 mm3 by inducing angiogenesis or through vasculogenic mimicry, the blood vessels composed of endothelial cells and tumor cells. However, vasculogenic mimicry forms blood vessels without endothelial cells. In fact, they are a mosaic blood vessel which allows substantial shedding of tumor cells into the circulation [7480]. Likewise, stromal proteins like VEGF and MMP-9 have been known to stimulate angiogenesis in PC [8183]. Consequently, it has been found that on an average metastatic cancer patient has in the vicinity of 5 and 50 CTCs for approximately every 7.5 ml of blood [8487]. Nonetheless, the number of CTCs varies between tumor types [88]. CTCs are found in both peripheral blood and bone marrow; additionally, CTCs in the bone marrow are generally called as disseminated tumor cells (DTCs) [8991]. Further, understanding the CTCs biological properties have demonstrated that the CTCs are involved in the distant organ colonization and metastatic spread of cancer [9296]. What's more, there has also been growing interest that mobilization of viable tumor cells from the site of primary tumor induced by a therapeutic or diagnostic intervention like surgery, radiation, chemotherapy or tissue biopsy may promote metastasis [97]. To comprehend this process, a team of researcher performed a tumor self-seeding mouse model experiment whereby tumor recurrence intervened by CTCs was examined utilizing human colorectal, melanoma and breast cancer cell lines, and found that tumor-derived cytokines IL-6 and IL-8 act as CTC attractants which were mediated by MMP1, FSCN1, and CXCL1 genes expressed on CTCs to promote infiltration [98]. However, it is not necessarily always associated with metastasis, as only 0.01% of CTCs takes part in metastasis [97, 99, 100]. Moreover, highly sensitive, single cell investigation showed marked heterogeneity of individual CTCs for protein expression and localization, and the CTCs reflected the character of both the primary biopsy and the transformations seen in the metastatic sites [101, 102], which corresponds with the evidence of “seed and soil” hypothesis [103105].

Amassing proof has demonstrated that CTCs can be utilized as a biomarker to non-invasively supervise cancer progression and provide direction to monitor the treatment [106108]. However, the American Society of Clinical Oncology (ASCO) fails to recommend the use of CTCs as a tumor marker for breast cancer, in the lack of strong evidence and conflicts of opinion between the experts [109, 110].

Nevertheless, CTCs detection, identification, enumeration, and molecular characterization are very challenging. Since, CTCs are uncommon in peripheral blood of patients (that is, 1–100 CTCs among billions of normal cells) with a half-life between 1 to 2.4 hours and due to its fragile nature, it tends to degrade when collected in standard blood collecting tubes [111116].

CTCs being a heterogeneous population of cells, CTCs can be positively or negatively enriched on the basis of 1. Physical characteristics and 2. Immunologic or biological characteristics. Physical features include; cell diameter >15 μm, nuclear-cytoplasmic ratio >0.8, electric charges, deformability, the hyperchromatic nuclei, sunken thickened nuclear membrane, and nucleus side-shift/large nucleoli/abnormal nuclear division. Whereas, cell surface protein expression, cell surface markers, RNA, and DNA signatures are used Immunologic or biological characteristics [117123]. The most common immunologic feature of CTCs exhibits, anti-CD45 antibody (for leukocytes), anti-vimentin antibody (mesenchymal marker), anti-CK8/18/19 antibody and the epithelial cell adhesion molecule (EpCAM) (epithelial marker) and contain a nucleus that binds to the nucleic acid dye 4’, 6-doamidino-2-phenylindole (DAPI) [117, 121]. Further, cytokeratin negative (CK-) CTCs are cancer stem cells (CSCs) that undergoes EMT. Moreover, CK-CTCs have a high potential for metastasis, and are the most resistant type of CTCs and importantly express genes associated with cancer [124, 125]. Similarly, CTCs that are undergoing apoptosis are called apoptotic CTCs, that can be detected by Epic Sciences technology that recognizes nuclear fragmentation or cytoplasmic blebbing associated with apoptosis, and measuring the ratio of viable CTCs and apoptotic CTCs may help in treatment monitoring [124, 126, 127].

Physical properties-based technologies such as ISET (isolation by size of epithelial tumor cells), ScreenCell, ApoStream™, density gradient centrifugation are used to detect, capture, and isolate CTCs. However, size-based selection strategies abuse the fact that CTCs are notably larger in size than normal blood cells [128, 129]. Importantly, nevertheless, this strategy probably results in substantial loss of CTCs which neglects the small CTCs that are cytokeratin positive and CD45 negative, and with similar sizes and shapes to white blood cells. Critically, these small CTCs have cancer-specific biomarkers that distinguish them as CTCs. Additionally, small CTCs have been found in dynamic illness and differentiation into small cell carcinomas, which often require an alternative treatment [130]. CTC cluster containing three or more CTCs is characterized as circulating tumor microemboli (CTM) [121, 129]. Moreover, CTM has high metastatic potential compared to single CTC [131133]. Lately, researchers involved in an animal experimental model found that a thrombolytic agent like urokinase can prevent the formation of CTM and further prolong overall survival approximately by 20% compared to control. Additionally, they also concluded that CTM mobilizes at a slower rate than the single CTC due to vessel wall adhesion [133].

In the recent years, several platforms have been established for segregation of CTCs that consider both positive and negative enrichment based on physical and immunological features consolidated within the same device [112, 118, 128, 129, 134147]. A complete outline of these strategies is beyond the scope of this article. Despite these many platforms, CellSearch™ remains only the gold standard and approved by FDA for all the CTCs detection strategies [148150]. In particular, this CellSearch™ strategy is dependent on the expression of epithelial markers by the CTCs, more specifically the Epithelial Cell Adhesion Molecule (EpCAM) [150, 151]. On the contrary, with respect to recent finding EpCAM based strategy fails to detect CTCs with low EpCAM expression and CK-CTCs, as CTCs tend to lose their epithelial antigens by the EMT process [152156]. In addition, it has also been recently revealed that EpCAM- negative CTCs are highly aggressive and invasive [154, 157]. Since CellSearch™ method is based on the idea that CTCs do not express the leukocyte antigen CD45, this method also neglects the fact that CTCs can directly attach to platelets and immune cells and thus accounts to be CD45-positive, which further evade immune surveillance that results in clonal expansion and metastatic [158]. In this manner, the CellSearch™ method may underestimate those CTCs that are highly aggressive and invasive [154].

Following detection and isolation of CTCs, the harvested tumor cells are studied for its genetic and biological features. Various molecular techniques, such as immunocytochemistry (ICC), fluorescence in situ hybridization (FISH), immunophenotyping, microarray, quantitative reverse transcription-polymerase chain reaction (qRT-PCR), droplet digital PCR (ddPCR), co-amplification at lower denaturation temperature-PCR (COLD-PCR), next-generation sequencing (NGS), beads, emulsion, amplification and magnetic (BEAMing), and whole genome amplification, to mention but a few have been commonly performed [159170]. Despite advancement in these molecular techniques, a genetic study of CTCs still faces challenges of sensitivity and specificity. In particular, digital PCR-based technology is able to screen genetic variations at a very low frequency of 0.01% [171]. However, it only permits monitoring of known mutations and limited numbers of foci [172175]. Bearing in mind, tumor cells change their mutation under the pressure of therapy or in the midst of tumor sub-clones; therefore, digital PCR-based technology may miss important information during the monitoring process. In addition to this, DNA sequencing of single CTCs for whole genome analysis requires obtaining adequate amounts of DNA and requires institutionalization [176178]. Moreover, for RNA sequencing strategies larger blood volume is required to obtain an adequate amount of CTCs. What's more, CTCs needs to be captured rapidly in order to avoid RNA degradation. Hence, it's not suitable for cancer screening at the moment [163].

To enhance sensitivity and specificity despite the heterogeneity of CTCs, innovative strategies have to be developed, that can consolidate physical, immunological and genetic analyses together to ease the detection, isolation, enrichment and molecular characterization of CTCs [179].

Potential clinical utility and research model of CTCs in PC

Currently, the clinical avail of CTCs analysis remains debatable in the PC. To date, numerous analysts have attempted to identify CTCs in patients with PC and have shown its potential clinical utility utilizing different methodologies and with varying results (Supplementary Table 1) [84, 180190]. Notably, some studies showed that CellSearch™ has a lower CTCs detection rate for PC patients with the sensitivity and specificity of 55.5 % and 100 %, respectively. Additionally, these studies also revealed that CTCs could only be found in malignant pancreatic tumor and CTCs positive patients have a significantly shorter overall survival. However, CTCs detected in these studies failed to correspond with tumor stage [84, 180, 182, 190]. Interestingly, in a study by Zhou et al. proposed that the integrated identification of c-Met, h-TERT, CK20, and CEA could be used as an indicator for CTCs in the circulation of a PC patient, which can be detected by combined use of immunomagnetic separation and RT-PCR, and thus, improving the specificity and sensitivity to 100%. Moreover, the positive expression of C-MET, CK20, and CEA was found to be closely correlated with tumor stage [189].

Ankeny et al. found that the numbers of CTCs detected from PC was able to differentiate different stages of disease as a useful biomarker and showed 100% similarity for KRAS mutation subtype between primary tumor and CTCs [187]. A meta-analysis comprising 623 patients with different stages of a PC revealed that the patients with positive CTCs had poor progress free survival (PFS) (HR=1.89, 95 %; CI=0.88–2.08, P<0.001) and overall survival (HR=1.23, 95 % CI=1.25–4.00, P<0.001) than those with the CTC-negative patients, suggesting CTCs may be a promising biomarker for the diagnosis and prognosis of a PC [191].

In a study by Yu et al., CTCs was isolated by the HbCTC-Chip microfluidic device from genetically engineered KPC mice and CTCs were subjected to single molecule RNA sequencing, they identified overexpression of the WNT2 gene in CTCs, which prevent anoikis, anchorage-independent sphere formation, and surge metastatic tendency in vivo. These findings were supported in CTCs investigated from 5 of 11 patients with PC. Thus, this study proposed that the molecular study of CTCs may recognize patient drug targets [192].

In spite of the fact that evidence indicates the abundance of tumor cells in the blood of patients with PC has prognostic value, and that CTC numbers can be used as a biomarker for diagnosis, staging of a PC before treatment, and can be prescient of response to therapy after treatment and, consequently, treatment results. These results must be considered with vigilance; however, because CTC numbers are highly variable between different CTC detection platforms, and are subject to favoritism relating to the variety of detection methods used. Thus, the quantity of CTCs that can be detected is therefore highly dependent on how the platform characterizes a cell as a CTC. Nevertheless, this impediment is likely to be overcome by consolidating different technologies to enhance analysis performance. Apart from the difference in results between CTCs detection platforms, some other hypothesis has been postulated for reasons behind low CTCs in the PC 1. The blood flow in the PC is notably compromised in contrast to that of the normal pancreas [193]. Thus, less number of CTCs shed into the circulation 2. The moderately low CTCs number reported in PC may be a consequence of CTCs sequestration by the liver as blood advances through the portal circulation into the systemic circulation [190, 194].

In contemporary research, the development of cell lines from CTCs is a motivating novel field. Recently, different groups reported CTCs developed cell lines in vitro from patients with breast, mesothelioma, esophageal, bladder, lung, and colon cancer [195200]. However, some researchers found it to be demanding and reported that CTCderived xenografts (CDXs) foresee therapeutic response conflictingly in many cancers, including the PC [195, 201204]. Interestingly, this has been confirmed by recent research that pluripotent stem cells cultured in the lab acquire new mutations all the time, especially in TP53 gene [205], and this might be a suggesting reason that why CDXs doesn't correspond to genetic mutations to that of the primary tumor, suggesting that research must be careful while genetic characterization of CDXs. By contrast, direct inoculation of CTCs into immune-compromised mice has met with appreciable success in lung cancer and showed to mirror the response of chemotherapy in CDXs model to that of donor patient. Additionally, it also exhibited comparable genetic profile between isolated CTCs and CDXs model [206]. CDX models have been developed for breast and prostate cancer [207209]. More interestingly, Gao and colleagues [210] successfully cultured 3D organoids system from CTCs isolated from patients with advanced prostate cancer, which they aim to recapitulate the different subtypes prostate cancer. Likewise, some group of researchers has successfully isolated CTCs from patient-derived tumor xeno-graft (PDTX) models in some cancers, including the PC, and identified its potential clinical utility [211213]. Perhaps, a standout amongst the most energizing utilizations of CTCs lines is that CDXs and organoids system model may bolster choice of targeted therapies, tracking cancer genetic and epigenetic modifications, and may evolve as an instrumental device for new drug development (Figure 1B). While initial studies using these models are promising, but it needs to be validated with further researches. The primary limitation of these models might be the selection method used for CTC enrichment. In a separate proof-of-concept study, Yu et al. created a pharmacogenomic (PGx) model to predict treatment response of a PC patient to chemotherapy regimens based on the genetic mutations in CTCs, and found that PGx profiling of CTCs can forecast treatment response, additionally they found clinical advantage for the patients treated with sensitive chemotherapy regimens versus insensitive chemotherapy regimens in regard to progression free (10.4 mo vs. 3.6 mo; P < 0.0001; HR, 0.14) and overall survival (17.2 mo vs. 8.3 mo; P < 0.0249; HR, 0.29) [214].

The big question remains: Can these CTCs derived xeno-graft and organoids models give a mirror image of a PC?

CIRCULATING TUMOR NUCLEIC ACIDS (CTNAS)

Circulating tumor nucleic acids (ctNAs) composed of circulating tumor DNA (ctDNA), mRNA and microRNA (miRNA) that are released and circulate in the blood of cancer patients, and changes in the levels of ctNAs in the circulation have been associated with tumor burden, tumor stage, vascularity, cellular turnover, response to therapy, and metastasis [17, 215217]. At present, the potential clinical utility of cell free RNA (cfRNA) is debatable. A comprehensive discussion of cfRNA is beyond the scope of this article, and this topic has been well documented elsewhere [218]. Moreover, miRNAs are most copious circulating RNA and are also carried in exosomes; thus, this topic has been covered later on with regards to exosomes in this article.

CIRCULATING TUMOR DNA (CTDNA)

It has been postulated that cell free DNA (cfDNA) can originate directly from the viable tumor cells or from CTCs by apoptosis, necrosis, autophagy, micro-environmental stress, mitotic catastrophe, trauma, and treatment procedure [17, 219226], others include viruses, such as EBV, HPV and hepatitis B virus [227229]. Moreover, cfDNA is regarded as ‘circulating tumor DNA’ (ctDNA) after mutations in cfDNA in cancer; hence, information regarding the origination and release of ctDNA may provide insight to clinicians about their possible involvement and nature of the disease. Of these, many studies have shown that ctDNA conveys genomic and epigenomic modifications indistinguishable to those of tumor cells [230]. Studies have demonstrated that cfDNA is cleared from the circulation by means of nuclease activity and renal clearance [231233]. Additionally, some cfDNA that are taken up by the liver and spleen are degraded by macrophages [234, 235]. Studies in both human and mice have shown that most of the apoptotic cfDNA fragments are measured in the vicinity of 166 and 200 base pairs (bp) with an observed half-life of 16 minutes to 2.5 hours [216, 236241]. In contrast, necrosis creates higher molecular weight DNA fragments of over 10,000 bp in size due to an inadequate and irregular absorption of genomic DNA [242245]. However, current isolation strategies are unable to capture long DNA fragments [246]. Indeed, the length of the cfDNA might be clinically valuable, utilized as a surrogate for identification of tumors as ctDNA released from necrosis represents malignant tumor's origin, and increased in the DNA integrity index (ratio of longer fragments to shorter DNA) are seen in most of the malignant tumors [247249]. However, there are clashing reports in the literature about the origin and composition of ctDNA, a few reports have shown that ctDNAs are shorter than that of apoptotic cfDNAs [238, 250, 251], measuring between 134 and 144 bp [250]. Withal, not all cfDNA originates from cell death; viable cells also release cfDNA as a part of homeostasis [221, 223, 252, 253]. In addition to this, it has also been seen that the activation of lymphocytes can result in the release of large numbers of cfDNA in the absence of apoptosis or necrosis [222, 252, 254]. Moreover, It has been suggested that cfDNA act as a ligand for Toll-like receptor 9 (TLR9) that may inhibit pro-apoptotic caspases by virtue of TLR9-dependent signaling [255]. This signifies a possible immunomodulatory function for cfDNA. These days cfDNA remains to be a hot topic and is widely used for a wide range of research and clinical purposes, including tumor genotyping, early cancer detection, patient prognosis, minimal residual disease monitoring, therapy evaluation, a biomarker in transplant surgery for graft injury, and prediction of allograft rejection [58, 256268]. In recent years, multiple studies have demonstrated that patients with invasive tumors such as lung, breast, pancreas, colon, hepatocellular, ovarian, prostate, esophageal, and melanoma generally have a high level of ctDNA in their plasma than in healthy individuals [269274]. Several genomic studies of tumor mutations have analyzed ctDNA to quantify the tumor burden and to detect therapeutic resistance conferring mutations [216, 275277]. Moreover, a correlation has been set up between the levels of non-mutated cfDNA and mutated cfDNA in circulation and the tumor stage [278, 279]. In addition to this, some studies have also found that mutated cfDNA can lead to therapeutic resistance in cancer several months prior to detection of the tumor by imaging, helping clinicians for therapy evaluation [275, 280, 281].

The study of ctDNA in the plasma basically involves quantification of ctDNA in the circulation using various measurements, for instance mutant allele fraction or mutant allele concentration (that is, copies per milliliter) to estimate disease burden and the detection of genetic aberrations such as somatic mutations, allelic imbalances, genetic polymorphisms, microsatellite alterations, loss of heterozygosity, and methylation [17, 256, 282288].

There are various methods and technologies used for quantitative and qualitative analysis of ctDNAs, commonly used platforms to name a few are digital PCR (dPCR) [289], droplet digital PCR (ddPCR) [290], BEAMing [291, 292], cancer personalized profiling by deep sequencing (CAPP-Seq) [293], tagged amplicon deep sequencing (TAM-Seq) [276], safe-sequencing (Safe-Seq) [294], duplex sequencing [295], integrated digital error suppression (iDES)-enhanced CAPP-Seq [296], whole-genome sequencing (WGS) [297, 298] and next-generation sequencing (NGS) [299]. Among these NGS holds great expectation for future of genomic analysis.

Regardless of these wide ranges of technologies, extraction, and analysis of ctDNA still face many challenges that need to be addressed before its regular use in a clinical setup. The major challenges are 1. Contamination of ctDNA with a large amount of wild-type cfDNA which are released from lysis of WBC of stored blood in EDTA tubes [300]. Hence, it has been proposed for the utilization of commercially available cell stabilization tubes to prevent or delay the lysis of WBC thereby decreasing the dilution impact of the ctDNA [301]. Additionally, a collection of blood at room temperature and shouldn't freeze more than 2 hours before extracting the plasma for ctDNA analysis, avoid of use heparinised tubes, extraction of ctDNA from plasma rather than serum and a double centrifugation step to remove more cellular debris preceding DNA extraction [300302]. 2. Low sensitivity and specificity for analysis of ctDNA [303, 304], this could be enhanced by the combination of advanced genomic approaches that have higher sensitivity to identify all ctDNA in the sample, even with small amounts of input material it has been found that multiplexed patient-specific panels in combination with targeted sequencing methods can improve the sensitivity [296, 304, 305]. 3. The expenses of NGS for liquid biopsies are high and the requirement for repeated liquid biopsies for longitudinal study may constrain its use among a substantial group of patients. The quick question emerges: Can patients bear the cost of advanced genomic approaches?

Potential clinical utility and research model of ctDNA in PC

ctDNA based liquid biopsy brings to the clinic the valuable strength for the success of targeted therapy and precision medicine. Straightaway, understanding of molecular landscapes of PC is vital to guide treatment decisions in clinical practice and with regards to clinical trials. Summary of selected studies using ctDNA for diagnosis, staging, and treatment of pancreatic cancer has been outlined in Table 1 [299, 306312]. It was Shapiro et al. who first reported the presence of ctDNA in PC and exhibited that ctDNA is markedly elevated in PC patients contrasted with healthy controls, and further concluded that ctDNA may serve as a useful diagnostic and prognostic biomarker [269]. Moreover, PC patients with noticeable ctDNA have been appearing to have worse survival and advanced disease stage [313]. In addition to this, ctDNA can be used for real-time monitoring of tumor dynamics, because of its short half-life it can present with the true picture of tumor burden in response to different therapy [216]. Sausen et al. demonstrated that the measurement of ctDNA can be used to predict relapse and poor outcome after curative surgery, and recurrence could be detected 6.5 months before radiographic imaging [310]. Moreover, in a recent study, ctDNA was found to be an independent prognostic marker in advanced PC, and the presence of ctDNA was associated with a shorter disease-free survival (4.6 vs.17.6 months) and overall survival (19.3 vs. 32.2 months) after surgery in patients with resectable PC [299]. In a separate study, Tjensvoll et al. noted that during chemotherapy of PC patients, changes in mutant KRAS gene level in the circulation corresponded with radiological imaging data and CA19-9 level; moreover, they proposed the utility of ctDNA for monitoring treatment efficacy and tumor progression [312]. These studies demonstrate the potential clinical utility of ctDNA as a prognostic biomarker in PC and further its benefit in monitoring minimal residual disease.

Table 1
Studies of circulating tumor DNA (ctDNA) in pancreatic cancer
StudyPts (N)StagesControlsTime of AnalysisPlatformMarkersFindingsSensitivity and Specificity
Zill et al. 2015 [309]18Advanced pancreatobiliary cancers8 biliary cancerPost-treatmentNGSKRAS, TP53, APC, SMAD4, and FBXW7Mutations were detected in 90.3% of cfDNA. The diagnostic accuracy of cfDNA sequencing was 97.7%, changes in cfDNA coordinated with tumor marker dynamics.92.3% and 100%
Cheng et al. 2017 [308]188Metastatic PCNAPre-treatmentNGS and ddPCRKRAS, BRCA2, EGFR and KDRThe KRAS mutation was detected in 72.3% (136/188) patients. The detection of ctDNA and response to treatment as assessed by CT imaging was 76.9%, the presence of ctDNA provided the earliest measure of treatment in 60% patients.NA
Berger et al. 2016 [307]24Metastatic PC21 IPMN, 38 healthy controls, 26 patients with resected SCAs and 16 borderline IPMNNAddPCRKRASThe KRAS mutation was detected in 41.7% (10/24) patients. KRAS mutation was not detected in cfDNA of controls, SCA, and IPMN.NA
Sausen et al. 2015 [310]77stage IINAPre-treatment and Post-treatmentNGS ddPCRNActDNA was detected in the 43% of patients with localized disease at diagnosis, and detection of ctDNA after resection predicts clinical relapse and poor prognosis. Moreover, ctDNA could detect recurrence 6.5 months earlier than with CT imaging.NA
Henriksen et al. 2016 [311]95NA27 without evidence of malignancy, 97 CP and 59 APNAEasyMag platform, PCRBMP3, RASSF1A, BNC1, MESTv2, TFPI2, APC, SFRP1 and SFRP2The distinction in mean number of methylated genes in the PC group (8.41 (95% CI 7.62–9.20)) versus the aggregate control group (4.74 (95% CI 4.40-5.08)) was highly significant (p < 0.001). Additionally, a diagnostic prediction model (age > 65, BMP3, RASSF1A, BNC1, MESTv2, TFPI2, APC, SFRP1 and SFRP2) had an area under the curve of 0.86 (sensitivity 76%, specificity 83%).NA
Tjensvoll et al. 2016 [312]14All stages29 healthy individualsPre-treatment and Post-treatmentPNA-clamp PCRKRASKRAS mutation was detected in 71% of patients with PC. The pre-therapy ctDNA was a predictor of both progression-free and OS. Changes in ctDNA levels corresponded both with radiological follow-up data and CA19-9 levels.NA
Maire et al. 2002 [306]47NA31 CPPre-treatment -PCR and allele-specific amplificationKRAS2KRAS2 mutation was detected in 22 patients (47%) with PC and in 4 controls with CP (13%) (P < 0.002). The combination of KRAS2 and CA19.9 gave a sensitivity and specificity of 98% and 77% respectively.47% and 87%
Pietrasz et al. 2017 [299]135All stagesNAPre-treatment and Post-treatmentNGSNActDNA was detected in 48% of patients with advanced PC, and ctDNA emerges as an independent prognostic marker in advanced PC and indicator of shorter DFS and shorter OS when detected after surgery.NA
PC: Pancreatic cancer; cfDNA: Cell-free DNA; PCR: Polymerase chain reaction; ddPCR: Droplet digital PCR; NGS: Next-generation sequencing; PNA-clamp PCR: Peptide nucleic acid clamping PCR; IPMN: Intraductal papillary mucinous neoplasm; SCAs: Serous cystadenomas; DFS: Disease-free survival OS: Overall survival; CP: Chronic pancreatitis; AP: Acute pancreatitis; CT: Computed tomography; ctDNA: Circulating tumor DNA; NA: Not available.

Since, it is known that over 90% of mutation in PC contains mutated KRAS gene and is considered to be an early event during carcinogenesis [8, 52, 314316]. Moreover, the mutation occurs most commonly in codon G12D, G12V, and G12R [314, 317, 318]. G12V mutation is significantly associated with shorter survival contrasted to G12D, G12R, and wild-type [319]. Thus, mutated KRAS gene has been a center of surveillance for definitive diagnosis of PC. Discouragingly, chronic pancreatitis also shows the KRAS mutation in 10 to 15% of the cases in cfDNA, and to increase the sensitivity and specificity for diagnosis of PC it has additionally been proposed that combining KRAS mutation and serum CA19-9 level can enhance a sensitivity and specificity by 98% and 77%, respectively [306]. Besides, later on, some studies have shown that methylation analysis of DNA can differentiate PC from chronic pancreatitis and could be used as a potential diagnostic marker for PC [311, 320, 321]. Additionally, it has been found that ctDNA methylation analysis can also detect epigenetic alterations in different cancers, including PC that involves in tumor progression and metastasis [322325]. Besides, these epigenetic alterations are strongly associated with patient survival [326]. Interestingly, recently it has been proposed that epigenetic alterations in a gene can be reprogrammed genetically or with a pharmacological inhibitor to reverse the epigenetic variations and inhibit their tumor-forming capacity; thus, helping in a finding of attractive therapeutic targets [327]. To the point, the essential question arises: Can ctDNA methylation analysis detect all the epigenetic alterations?

In addition to this, methylation of ctDNA has been found to conceal tissue and cell specific information that may be invaluable in cancer patients to find tissue-of-origin [251, 328], such as in the case of cancers of unknown origin. Of note, methylation analysis of ctDNA is found to be useful in determining the primary location of cancer with a specificity and sensitivity of 99.6% and 97.7%, respectively [329].

Apart from KRAS mutation, in recent years with the development of NGS, increasingly pertinent genetic aberrations have been identified, namely oncogenic BRAF V600E mutation that are observed 3% of PC patients, and that do not acquire a KRAS mutation; similarly, amplification of the MYC oncogene which is remarkably associated with poor prognosis [318]. Moreover, detection of mutations such as RBM10, MLL, MLL2, MLL3, and ARID1A is associated with longer survival [310, 318]. These studies furnish genetic indicators of prognosis and outcome in PC and have suggestions for a new period of therapeutic development (Figure 1C).

In a separate study carried by Zill et al. has demonstrated that tumor sequencing was failed in 35% cases of tissue biopsy due to inadequate a tissue sample, in addition to this all mutations were detected in ctDNA similar to that of tissue biopsy. Moreover, they proposed that ctDNA could correlate well with tumor marker dynamics in longitudinal monitoring with a diagnostic accuracy of 97.7%, and with sensitivity and specificity of 92.3% and 100%, respectively [309].

Intra- and inter-tumor heterogeneity contribute to the development of drug-resistant tumors and failure of treatment [330]. A small genetic clone carrying a drug-resistant mutation within the tumor can extend after the pressure treatment. These genetic clones can be missed by tissue biopsies due to low prevalence or the spatial partition of cells within the tumor [331]. Interestingly, ctDNA can be exploited to monitor dynamic clonal and subclonal evolution in response to the pressure of therapy [332] (Figure 2).

Figure 2

Tumor heterogeneity and clonal evolution during treatment

(A) Diagram showing the evolutional clonal architecture in pancreatic cancer (PC) at diagnosis and relapse. Of note, at diagnosis, the clonal and subclonal diversity evolved from a common ancestral tumor stem cell. The clonal evolution may follow linear or branched evolution, however, branched evolution is probably more likely to contribute to tumor heterogeneity. Additionally, drug treatment instigates a bottleneck effect, where resistant subclones will survive and proliferate to form a heterogeneous tumor. (B) During systemic successive targeted therapy assessed by longitudinal liquid biopsies may identify an actionable genetic alteration, therapy response or progression. In the event that progression is identified, the clinician may be able to switch treatment to target arising clones that carry additional mutations that were identified by the ctDNA analysis. At the start of targeted therapy, all cells in the patient's with PC have actionable genetic mutations (clone 1). The administration of treatment 1 targets the clone 1. longitudinal liquid biopsy analysis demonstrates an initial decrease in the clone 1 during treatment 1, yet uncovers the evolution of new clone (clone 2 and clone 3) causing resistance to treatment 1. The clone 2 and clone 3 can be targeted with treatment 2, where longitudinal liquid biopsy analysis uncovers a decrease in the frequency of resistance clone 2 and clone 3, during this time, however, other genetic alterations clone 4 and clone 5 increases in frequency. These clones 4 and 5 are resistant to treatment 2, yet is sensitive to treatment 3. During treatment 3, the frequency of the clone 4 and clone 5 decreases, while residual earlier resistant clones may persist to give rise to therapeutic resistance.

EXOSOMES

Exosomes are very stable, small cup-shaped, lipid bilayer microvesicles of endocytic origin with a size of 50–150 nm in diameter and density of 1.12–1.19 g/ml [60, 333, 334]. These microvesicles are discharged by all cells, including tumor cells, and originally thought to be that they are like cellular garbage bags [335337]. However, recent research suggests that exosomes are involved in many physiological and pathological functions and processes such as intracellular communication, inflammation, cell proliferation and regeneration following injury, immune response, lactation, neuronal function, immunothrombosis, diabetes, atherosclerosis, development, and progression of liver disease, neurodegenerative diseases and more recently in cancer [338352]. Evidence demonstrates that exosomes are available in numerous biologic body fluids; exosomes may in this way be viewed as open indicative biomarkers that hold incredible potential for recognition of numerous disease conditions, including cancer [353, 354]. Interestingly, exosomes are enriched with DNA, proteins, lipids, RNAs, and metabolites that are reflective of the cell types of origin [352, 355357]. Nonetheless, whole RNA sequence can't be bundled inside one exosome, because of its small size, which was contrasted and retrovirus particles of a comparable size that can just pack 10 kb transcriptome, subsequently, single exosome conveys just a predetermined number of transcripts [358]. Still, exosomes are remarkably abundant in plasma and when segregating the vesicle portion, the vast majority of the RNA sequence can be identified [359, 360]. Recent data from various cell type uncovers that exosomes contain 4,563 proteins, 194 lipids, 1639 mRNA and 764 microRNA [361]. Among the main 20 regularly recognized exosomal proteins, a significant number of the proteins, including CD9, ACTB, CD63, CD81, HSPA8, PKM2, ANXA2, HSP90A1, SDCBP, YWHAE, LDHA, MSN, PDCP6IP, ANXA5, FASN, ACTN4, LDHB, ANXA1, HSPA1A, and YWHA are known to be mutated in multiple cancer types [362]. Additionally, it has been found that some exosomes reveal major histocompatibility complex MHC I and MHC II on their surface, suggesting that they are derived from antigen-presenting cells and might have a regulatory immunological part in cancer biology [363, 364]. Considering, exosomes convey genomic and proteomic materials, thus it has been hypothesized that exosomes secreted by tumor cells take part in the tumor growth, invasion, pre-metastatic niches (PMNs) and metastasis through intracellular communication and escape from immunosurveillance [338, 365373]. Biogenesis and secretion of exosome within the cell is a complex process which requires different factors like molecular motors (cytoskeleton- kinesins and myosins, polymerisation- actin, dynamin, and microtubules), molecular switches (GTPases, annexins, and flotillin) and the fusion proteins (SNARE proteins and tethering factors), cargo sorting proteins complex (ALIX and TSG101) and finally exocytotic released is promoted by cellular stress or hypoxia in cancer cells [333, 374377]. Studies from Thery and colleagues reveal that Rab27a and Rab27b act as key downregulators of the exosome secretion pathway, which inhibit secretion of exosomes [378]. These Rab family proteins are thought to be involved in cancer progression and tumor advancement, which provided clues that exosomes have something to do with tumor biology [379]. Moreover, p53 protein, and a p53-regulated gene, TSAP6, have shown to increase production of exosome [380, 381]. Emerging evidence suggested that breast and pancreatic tumor-derived exosomes express integrins (ITGs) on their surface which direct organ-specific colonization by fusing with targeted stromal and cancer cells, thereby forming PMNs within the cancer microenvironment to transmit signals and their cargo, that includes genetic material (that is, DNA, mRNA, and miRNA), metabolites and proteins, by that determining organotropic metastasis [338, 366368, 373, 382384]. Subsequently, PMNs requires S100 family proteins for homing of tumor-derived exosomes in targeted organs [373]. Furthermore, tumor-derived exosomal miRNA and protein have a tendency to reprogram and instruct target cells that it fuse with towards pro-inflammatory and pro-metastatic phenotype leading to metastasis [382, 385].

Till the date, numerous of technologies and methods have been used for extraction of exosomes from body fluids which have been well documented elsewhere [386]. Commonly used methods are ultracentrifugation-based isolation, precipitation-based isolation, size-based isolation, immunoaffinity-based isolation, and microfluidics-based isolation [386]. The segregation of tumor-derived exosomes from patients; however, remains challenging due to some of the reasons: 1. Lack of a standardized method for segregation and the absence of specific markers that can differentiate tumor-derived and non-tumor derived exosomes [387]. 2. Failure in isolating large concentration of exosomes, due to contamination from other extracellular vesicles and cellular debris [386]. 3. Time-consuming technology that is hard to implement in routine clinical setup [386].

To overcome these challenges, an institutionalized technique for exosome isolation should be developed sooner rather than later, thus amplifying the significance of research facility based investigations of exosomes in the clinical setting. It is important to reliably approve each of these strategies as per meticulous definitions of exosomes, those laid out by the International Society for Extracellular Vesicles [388].

Potential clinical utility and research model of exosomes in PC

As the content of exosomes is cell-type specific with an extensive variety of molecular information carried forth from parent cells to secondary cells, exosomes may provide an idiosyncratic ‘signature’ of tumor development and metastatic progression, as well as the metabolic status of the tumor. In spite of the fact, that the mechanism of packaging is yet to be completely comprehended, it has been seen that the metastatic tumor cells show the high ability of packing and cargo secretion (that is, protein, RNA, DNA, and metabolites) in exosome [382, 385]. To date, numerous studies have outlined clinical utility of exosomes as a diagnostic, prognostic and therapeutic tool in PC patients (Table 2) [387, 389393].

Table 2
Studies of circulating tumor exosomes in pancreatic cancer
StudySpecimen typePlatformMarkersFindings and Conclusion
Que et al. 2013 [389]SerumFiltration, Ultracentrifugation, and RT-PCRmiR-17-5p, miR-21, miR-155, and miR-196aThere were low expressions of exosomal miR-155 and miR-196a in PC patients. Moreover, there were high expressions of serum exosomal miR-17-5p and miR-21 in PC patients than control groups and high expression of miR-17-5p was significantly correlated with advanced stage of PC.
Kahlert et al. 2014 [390]SerumFiltration, Ultracentrifugation and WGSKRAS, p53Exosomes from PC patients contain >10-kb fragments of double-stranded genomic DNA with detectable mutations in KRAS and p53. In addition, WGS of exosomal DNA can determine genomic DNA mutations for cancer prediction, treatment, and therapy resistance.
Madhavan et al. 2015 [391]SerumUltracentrifugation, qRT-PCR, and Flow cytometryCD44v6, Tspan8, EpCAM, c-Met, CD104, miR-1246, miR-4644, miR-3976, and miR-4306Serum exosomal miR-1246, miR-4644, miR-3976 and miR-4306 were significantly upregulated in 83% of PC serum-exosomes, but rarely in control groups. Additionally, It was found that a combination of five proteins (CD44v6, Tspan8, EpCAM, MET and CD104) and four miRNAs (miR-1246, miR-4644, miR-3976 and miR-4306) in circulating tumor exosomes could recognize PC from healthy control, chronic pancreatitis and benign pancreatic disease with a sensitivity and specificity of 100% and 80% respectively.
Melo et al. 2015 [387]SerumFiltration, Ultracentrifugation, qRT-PCR And Mass spectrometry analysesGlypican-1Expression of glypican 1 (GPC1) a membrane-bound protein on circulating exosomes of mice and humans with PC can differentiate healthy control and patients with a benign pancreatic disease. Notably, GPC1+ exosomes level correlated with tumor burden and the survival of PC patients before and after the surgery with utter sensitivity and specificity. Additionally, circulating GPC1+ exosomes of PC patients bear KRAS mutations,
Kanwar et al. 2014 [392]SerumExoChip (antigen based)CD63Significantly higher exosome capture in PC patients, compared to controls.
Allenson et al. 2017 [393]Whole bloodUltracentrifugation, Flow cytometry, and ddPCRKRASExosomal DNA posses KRAS mutations and was detected localized, locally advanced, and metastatic PC patients, respectively. Higher exosomal DNA KRAS mutations were associated with decreased disease-free survival in patients with localized disease.
PC: pancreatic cancer; GPC1: glypican-1; miR: microRNA; ddPCR: Droplet digital polymerase chain reaction; qRT-PCR: Quantitative reverse transcription polymerase chain reaction; WGS: Whole-genome sequencing; EpCAM: Epithelial cellular adhesion molecule; RT-PCR: Reverse transcription polymerase chain reaction.

In a seminal research, Melo et al. [387] demonstrated an increased amount of glypican 1 (GPC1) a membrane-bound protein on circulating exosomes of mice and humans with PC can differentiate healthy control and patients with a benign pancreatic disease. Notably, GPC1+ exosomes level correlated with tumor burden and the survival of PC patients before and after the surgery with utter sensitivity and specificity. Additionally, circulating GPC1+ exosomes of PC patients bear KRAS mutations, and were able to identify pancreatic intraepithelial neoplasia (PanIN) in mice from healthy control even before detectable pancreatic lesion on MRI. Of note, the main limitation of this study was a small sample size. Undoubtedly, these findings should be verified with a larger series of the sample, but the striking evidence provided by Melo and colleagues suggest that GPC1+ exosomes may serve as a potential diagnostic and screening biomarker to detect early stages of PC for possible curative surgery. In earlier studies, overexpression of surviving [394], and mislocalization of plectin [395] in exosomes were also proposed as biomarkers for PC. Moreover, it has also recently been found that a higher rate of patients with localized PC showed noticeable KRAS mutations in exosomal DNA than previously revealed for cfDNA, and thus exosomal DNA may act as a complementary DNA source to liquid biopsy [393]. In a research, Madhavan et al. outlined that a combination of five proteins (CD44v6, Tspan8, EpCAM, MET and CD104) and four miRNAs (miR-1246, miR-4644, miR-3976 and miR-4306) in circulating tumor exosomes could recognize PC from healthy control, chronic pancreatitis, and benign pancreatic disease with a sensitivity and specificity of 100% and 80%, respectively [391].

Exosomal micro-RNAs (miRNAs) have additionally increased generous consideration in later past years. From the recent studies, the number of exosomal miRNAs including miR-21, miR-17-5p, miR-155, miR-34, miR-196a, miR-181a, miR-181b, miR-138-5p, miR-494, miR-542-3p, miR-31, and miR-205 has been identified and upregulation of these miRNAs has been shown to increase cellular proliferation, angiogenesis promotion, disease progression, metastasis, and chemo-resistance in PC patients [389, 396403]. Moreover, these studies highlight the potential use of exosomal miRNAs as a diagnostic and prognostic biomarker. Likewise, targeting the exosomal miRNAs might be a potential therapy for PC.

Additionally, it has been found that miRNAs in circulating exosomes are representative of those increased in the primary tumor cells [21]. In a separate study, Ohuchida et al. distinguished 24 miRNAs with altered expression in gemcitabine-resistant cells, and furthermore found that patients with high miR-142-5p and miR-204 expression had significantly longer survival times than those with low miR-142-5p and miR-204 expression in the gemcitabine-treated group [404]. Despite the fact that the miRNA levels were determined in paraffin-embedded tissue, this highlights the potential use of tumor-derived exosomal miRNAs as predictors of response to chemotherapy and future use of miRNAs for targeted immune therapy in PC. Moreover, it had been proposed that exosomes miRNAs are derived from living cells, while circulating free miRNAs usually originates from apoptotic or necrotic cells [398], and thus, exosomes miRNAs might have advantages over circulating free miRNAs for monitoring therapy or late stage of PC.

Lyden and colleagues [373] in their recent paper proposed that tumor-derived exosomes integrins (ITGs) can determine organotropic metastasis as discussed in the earlier section of this paper. The consequent analysis demonstrated that liver-tropic pancreatic exosomes expressing ITGαvβ5 could communicate with F4/80+ macrophages and fuse with Kupffer cells in fibronectin rich liver niches. Besides, inhibiting ITGβ5 expression through short hairpin RNAs or hindering their binding by HYD-1/RGD peptides particularly reduced exosome uptake and additionally liver metastasis. This study explains why the liver is the most common site for PC metastasis. Moreover, these outcomes showed that exosomal ITGs may be used as organotropic biomarkers to anticipate organ-specific metastasis in PC patients, and expands our understanding of the organ-specific metastasis mechanisms involvement of exosomes in advancing tumor metastasis. In addition to Lyden results, a study by Costa-Silva et al. [385] demonstrated that the PC exosomes can expand liver metastatic burden by transferring macrophage migration inhibitory factor (MIF) to Kupffer cells and by recruiting immune cells to initiate PMNs development in the liver. It is thus proposed that the presence of MIF in exosomes may be a biomarker that can show the likelihood of PC metastasis to the liver and blockage of MIF could prevent liver metastases and may prove to be clinically relevant for the development of new targeted therapies.

The utilization of exosomes as a nucleic acid, gene or drug delivery vehicles (Figure 1D) has increased significant enthusiasm because of their phenomenal biodistribution and biocompatibility [405]. Moreover, the advantage of utilizing exosomes as a drug delivery system lies in the fact that they can be particularly targeted to a specific cell type by engineering exosome-producer cells [406]. Interestingly, past studies have demonstrated that tumor cells secrete more exosomes compared to normal cells [407]. Furthermore, malignant pancreatic tumor cells, with oncogenic RAS have also founded to uptake exosomes more readily through the active induction of macropinocytosis [408, 409], and this could strengthen the use of exosome as an ideal drug delivery vehicle. Recently, Kamerkar et al. demonstrated treatment efficacy of engineered exosome (iExosome), where iExosome was able to suppress the PC progression in genetically engineered KTC and KPC mouse models, this study exhibited an approach for direct and specific targeting of KRAS mutation in tumors using engineered exosomes [410]. However, it still needs to be verified in the clinical setting.

CONCLUSIONS

We have accomplished enormous progress in our understanding of the complex molecular and genetic mechanisms of PC, yet key inquiries stay unanswered for its early diagnosis, staging, treatment monitoring, and management. Taking everything into account, the up and coming era of ‘ liquid biopsy’ will be vital to conclusively build up the clinical relevance of bloodbased genomic profiling. Liquid biopsy methodologies will most likely give enhanced diagnostic and therapeutic outcome. However, a few issues stay to be tackled before application in a clinical setting: 1. Institutionalization of the sample collection methodology in pre-analytical setup, subsequently decreasing pre-analytical errors 2. Institutionalized and strict definition of CTCs, ctDNA, and exosomes for their segregation and analysis is required. 3. Improvement in the sensitivity and specificity of the detection methods by integrating CTCs, ctDNA, and exosomes in one platform. 4. Universal signature from CTCs, ctDNA, and exosomes for differentiating benign from malignant disease and that can cover all phases of cancer along with their subtypes, tumor characteristics, and mutations for the success of precision medicine. 5. Substantial forthcoming clinical trials, including multicenter studies, are expected to approve the clinical essentials for diagnosis, treatment monitoring, and prognosis. The comparison of CTCs, ctDNA, and exosomes is outlined in Table 3.

Table 3
Comparison between CTCs, ctDNA and exosomes as liquid biopsy
ComparisonCTCsctDNAExosomes
OriginIncludes apoptotic tumor and viable tumor cells from a primary or metastatic tumor [71, 73].Includes cfDNA, from the viable tumor cells or from CTCs [17, 219, 221, 224, 225].Includes DNA, proteins, lipids, RNAs and metabolites and are discharged by all cells including tumor cells [333, 336, 352, 355357].
Bio-banked samples used for studyFrozen plasma, urine and other biofluids cannot be used for study of CTCs [116].Frozen plasma, urine and other biofluids can be used for study of cfDNA [116, 260].Frozen plasma, urine and other biofluids can be used for study of exosomes [260].
StabilityUnstable [114, 116, 117]Stable [116]Very stable [334]
Genetic materialsDNA and RNA [117, 122, 123]DNA [17, 219, 221]DNA and RNA [355, 357, 359361]
Analytic TechniquesCellSearch [84, 148150, 180, 182, 190], Microfluidic [187, 181], SE-iFISH [186], MetaCell [184], Immunofluorescence [181, 185, 188], ScreenCell [185], ISET Test [188, 190] etc.dPCR [289], CAPP-Seq [293], TAM-Seq [276], ddPCR [290, 307, 308, 310], COLD-PCR [167], Safe-Seq [294], NGS [299, 308310], BEAMing [291, 292], WGA [297, 298] etc.Ultracentrifugation [387, 386, 389391, 393], ExoChip [392], Precipitation [386], Size-based isolation [386], Immunoaffinity-based isolation [386], Microfluidics-based isolation etc [386].
Morphological study and functional study of tumor cells ex vivoYes [128, 129, 188, 190, 206, 210, 213, 214]NoNo
Analysis of protein location on tumor cellsYes [101, 102]NoNo
Identification of mutationsYes [165, 185, 187]Yes [306, 319]Yes [387, 390, 393]
Identification of epigenetic changesYes [414, 415]Yes [322325]Yes [357]
Analysis of RNA transcription profilesYes [157, 192]NoYes [359361, 367]
Proteomics AnalysisYes [157]NoYes [367, 387, 392]
Analysis tumor heterogeneityYes [206]Yes [332]No
Use as drug delivery vehicleNoNoYes [406, 410]

DNA: Deoxyribonucleic acid; RNA: Ribonucleic acid; CTCs: Circulating tumor cells; ctDNA: Circulating tumor DNA; cfDNA: Cell free DNA; ISET: Size of Epithelial Tumor; SE-iFISH: Immunostaining-fluorescence in situ hybridization; ddPCR: Droplet digital PCR; COLD-PCR: Co-amplifcation at lower denaturation temperature-PCR; NGS: Next-generation sequencing; BEAMing: Beads, Emulsion, Amplifcation and Magnetic; WGA: Whole genome amplifications, messenger RNAs (mRNAs); dPCR: digital PCR; CAPP-Seq: Cancer personalized profiling by deep sequencin; TAM-Seq: Tagged amplicon deep sequencing; Safe-Seq: Safe-sequencing.

In spite of the presence of various challenges, liquid biopsy seems to be ideal diagnostic and therapeutic strategies for PC. So far, in June 2016, a liquid biopsy was approved by the FDA for use in the USA to detect EGFR mutations in plasma ctDNA and entered clinical practice for the management of non-small cell lung cancer (NSCLC) [68]. After its approval, it represents key milestones towards the application of liquid biopsies in personalized clinical oncology.

CTCs seem to have enormous potential for PC, and can be exploited to understand the development of the distant organ colonization and metastatic spread of cancer. Moreover, CTCs can be used to understand the phenotypic changes, plasticity of tumor biology and mutational landscape of tumor by development of PDTX [213], PGx [214], CDXs [206], and 3D organoids [210] models and guide treatment decisions for complex disease like PC [184, 190, 411413]. However, methylation analysis of CTCs remains largely unexplored, except few studies which have been reported in recent years [414, 415]. Methylation study of CTCs holds a promising future with an exciting result; this may give a new direction to upcoming research.

In the ctDNA arena, ctDNA has offered more an inclusive understanding of a patient's disease. For instance, the total ctDNA concentration can be used for real-time monitoring of tumor dynamics and predicts relapse, poor outcome and shorter disease-free survival after curative surgery [299, 310, 313]. Methylation analysis of ctDNA can detect epigenetic alterations that involve in tumor progression and metastasis [322, 325]. Moreover, it can also differentiate PC from chronic pancreatitis and could be used as a potential diagnostic marker for PC [320, 321]. However, the sensitivity and specificity of ctDNA analysis are struggling and it can be increased by adopting a multi-marker strategy along with integrating it with other biomarkers.

Exosomes provide an enormous understanding about organ-specific colonization and PMNs [373, 385]. Moreover, exosomes can serve as a potential biomarker as its contents are largely derived from the tumors, which are enriched with DNA, proteins, lipids, RNAs, and metabolites. In addition to this, exosomes as drug delivery vehicles offer an important perspective because of its cell-specific nature, excellent biodistribution and biocompatibility [405]. Before these drug delivery systems become a therapeutic reality, it needs to be validated with further researches and large clinical trials.

In general, a liquid biopsy can possibly be used to diagnose PC at an early stage, predict prognosis, monitor PC stage, therapeutic efficacy or resistance, and provide optimal, personalized treatment strategies for patients with PC. This review has endeavored to organize the present advances in liquid biopsy for PC into a solitary idea to establish an effective management plan and implementation of these understandings to bolster energizing zones of research. But the fundamental question remains: Can liquid biopsy become a screening reality for pancreatic cancer?

SUPPLEMENTARY MATERIALS TABLES

Footnotes

CONFLICTS OF INTEREST

Authors declare no competing interests.

FUNDING

This work was supported by grants from - The National High Technology Research and Development Program of China (SS2015AA020405); Training Program of the Key Program of the National Natural Science Foundation of China (91442115); The National Natural Science Foundation of China (No.81672337); Zhejiang Provincial Key Innovation Team of Pancreatic Cancer Diagnosis & Treatment (2013TD06); The Key Program of the National Natural Science Foundation of China (81530079); Key research and development Project of Zhejiang Province (2015C03044) (Sponsored by Zhejiang Provincial Program for the Cultivation of High-level Innovative Health talents); Zhejiang Provincial Key Laboratory of Pancreatic Disease; and National High Technology Research and Development Program of China (2015AA020405).

References

  • 1. CourtCMAnkenyJSHouSTsengHRTomlinsonJSImproving pancreatic cancer diagnosis using circulating tumor cells: prospects for staging and single-cell analysisExpert Rev Mol Diagn2015151491504https://doi.org/10.1586/14737159.2015.1091311[PubMed][Google Scholar]
  • 2. SiegelRLMillerKDJemalACancer Statistics, 2017CA Cancer J Clin201767730https://doi.org/10.3322/caac.21387[PubMed][Google Scholar]
  • 3. ChenWZhengRBaadePDZhangSZengHBrayFJemalAYuXQHeJCancer statistics in China, 2015CA Cancer J Clin20166611532https://doi.org/10.3322/caac.21338[PubMed][Google Scholar]
  • 4. AssifiMMLuXEiblGReberHALiGHinesOJNeoadjuvant therapy in pancreatic adenocarcinoma: a meta-analysis of phase II trialsSurgery201115046673https://doi.org/10.1016/j.surg.2011.07.006[PubMed][Google Scholar]
  • 5. ShrikhandeSVKleeffJReiserCWeitzJHinzUEspositoISchmidtJFriessHBuchlerMWPancreatic resection for M1 pancreatic ductal adenocarcinomaAnn Surg Oncol20071411827https://doi.org/10.1245/s10434-006-9131-8[PubMed][Google Scholar]
  • 6. YachidaSJonesSBozicIAntalTLearyRFuBKamiyamaMHrubanRHEshlemanJRNowakMAVelculescuVEKinzlerKWVogelsteinBDistant metastasis occurs late during the genetic evolution of pancreatic cancerNature201046711147https://doi.org/10.1038/nature09515[PubMed][Google Scholar]
  • 7. RahibLFleshmanJMMatrisianLMBerlinJDEvaluation of Pancreatic Cancer Clinical Trials and Benchmarks for Clinically Meaningful Future Trials: A Systematic ReviewJAMA Oncol20162120916https://doi.org/10.1001/jamaoncol.2016.0585[PubMed][Google Scholar]
  • 8. BaileyPChangDKNonesKJohnsALPatchAMGingrasMCMillerDKChristANBruxnerTJQuinnMCNourseCMurtaughLCHarliwongIGenomic analyses identify molecular subtypes of pancreatic cancerNature20165314752https://doi.org/10.1038/nature16965[PubMed][Google Scholar]
  • 9. HanahanDWeinbergRAHallmarks of cancer: the next generationCell201114464674https://doi.org/10.1016/j.cell.2011.02.013[PubMed][Google Scholar]
  • 10. HanahanDWeinbergRAThe hallmarks of cancerCell20001005770[PubMed][Google Scholar]
  • 11. MaitraAHrubanRHPancreatic cancerAnnu Rev Pathol2008315788https://doi.org/10.1146/annurev.pathmechdis.3.121806.154305[PubMed][Google Scholar]
  • 12. MimeaultMBrandRESassonAABatraSKRecent advances on the molecular mechanisms involved in pancreatic cancer progression and therapiesPancreas20053130116[PubMed][Google Scholar]
  • 13. CollissonEASadanandamAOlsonPGibbWJTruittMGuSCoocJWeinkleJKimGEJakkulaLFeilerHSKoAHOlshenABSubtypes of pancreatic ductal adenocarcinoma and their differing responses to therapyNat Med2011175003https://doi.org/10.1038/nm.2344[PubMed][Google Scholar]
  • 14. MoffittRAMarayatiRFlateELVolmarKELoezaSGHoadleyKARashidNUWilliamsLAEatonSCChungAHSmylaJKAndersonJMKimHJVirtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinomaNat Genet201547116878https://doi.org/10.1038/ng.3398[PubMed][Google Scholar]
  • 15. WaddellNPajicMPatchAMChangDKKassahnKSBaileyPJohnsALMillerDNonesKQuekKQuinnMCRobertsonAJFadlullahMZWhole genomes redefine the mutational landscape of pancreatic cancerNature2015518495501https://doi.org/10.1038/nature14169[PubMed][Google Scholar]
  • 16. Alix-PanabieresCPantelKCirculating tumor cells: liquid biopsy of cancerClin Chem2013591108https://doi.org/10.1373/clinchem.2012.194258[PubMed][Google Scholar]
  • 17. SchwarzenbachHHoonDSBPantelKCell-free nucleic acids as biomarkers in cancer patientsNat Rev Cancer20111142637[PubMed][Google Scholar]
  • 18. van de StolpeAPantelKSleijferSTerstappenLWden ToonderJMCirculating tumor cell isolation and diagnostics: toward routine clinical useCancer Res201171595560https://doi.org/10.1158/0008-5472.can-11-1254[PubMed][Google Scholar]
  • 19. CrowleyEDi NicolantonioFLoupakisFBardelliALiquid biopsy: monitoring cancer-genetics in the bloodNat Rev Clin Oncol20131047284https://doi.org/10.1038/nrclinonc.2013.110[PubMed][Google Scholar]
  • 20. GoldBCankovicMFurtadoLVMeierFGockeCDDo Circulating Tumor Cells, Exosomes, and Circulating Tumor Nucleic Acids Have Clinical Utility?: A Report of the Association for Molecular PathologyJ Mol Diagn20151720924https://doi.org/10.1016/j.jmoldx.2015.02.001[PubMed][Google Scholar]
  • 21. NedaeiniaRManianMJazayeriMHRanjbarMSalehiRSharifiMMohagheghFGoliMJahedniaSHAvanAGhayour-MobarhanMCirculating exosomes and exosomal microRNAs as biomarkers in gastrointestinal cancerCancer Gene Ther2017244856https://doi.org/10.1038/cgt.2016.77[PubMed][Google Scholar]
  • 22. AnkerPMulcahyHQi ChenXStrounMDetection of Circulating Tumour DNA in the Blood (Plasma/Serum) of Cancer PatientsCancer Metastasis Rev1999186573https://doi.org/10.1023/a:1006260319913[PubMed][Google Scholar]
  • 23. KimMSZhongYYachidaSRajeshkumarNVAbelMLMarimuthuAMudgalKHrubanRHPolingJSTynerJWMaitraAIacobuzio-DonahueCAPandeyAHeterogeneity of pancreatic cancer metastases in a single patient revealed by quantitative proteomicsMol Cell Proteomics201413280311https://doi.org/10.1074/mcp.M114.038547[PubMed][Google Scholar]
  • 24. CampbellPJYachidaSMudieLJStephensPJPleasanceEDStebbingsLAMorsbergerLALatimerCMcLarenSLinMLMcBrideDJVarelaINik-ZainalSAThe patterns and dynamics of genomic instability in metastatic pancreatic cancerNature2010467110913https://doi.org/10.1038/nature09460[PubMed][Google Scholar]
  • 25. LinQJYangFJinCFuDLCurrent status and progress of pancreatic cancer in ChinaWorld J Gastroenterol20152179888003https://doi.org/10.3748/wjg.v21.i26.7988[PubMed][Google Scholar]
  • 26. Pancreatic adenocarcinoma guidelinesNational Cancer Comprehensive Network2017Version 2.2016
  • 27. CaoSHuYGaoXLiaoQZhaoYSerum Carbohydrate Antigen 19-9 in Differential Diagnosis of Benign and Malignant Pancreatic Cystic Neoplasms: A Meta-AnalysisPLoS One201611e0166406https://doi.org/10.1371/journal.pone.0166406[PubMed][Google Scholar]
  • 28. WinterJMCameronJLCampbellKAArnoldMAChangDCColemanJHodginMBSauterPKHrubanRHRiallTSSchulickRDChotiMALillemoeKD1423 pancreaticoduodenectomies for pancreatic cancer: A single-institution experienceJ Gastrointest Surg2006101199210https://doi.org/10.1016/j.gassur.2006.08.018[PubMed][Google Scholar]
  • 29. KimJELeeKTLeeJKPaikSWRheeJCChoiKWClinical usefulness of carbohydrate antigen 19-9 as a screening test for pancreatic cancer in an asymptomatic populationJ Gastroenterol Hepatol2004191826[PubMed][Google Scholar]
  • 30. HommaTTsuchiyaRThe study of the mass screening of persons without symptoms and of the screening of outpatients with gastrointestinal complaints or icterus for pancreatic cancer in Japan, using CA19-9 and elastase-1 or ultrasonographyInt J Pancreatol1991911924[PubMed][Google Scholar]
  • 31. KarmazanovskyGFedorovVKubyshkinVKotchatkovAPancreatic head cancer: accuracy of CT in determination of resectabilityAbdom Imaging200530488500https://doi.org/10.1007/s00261-004-0279-z[PubMed][Google Scholar]
  • 32. AppelBLTolatPEvansDBTsaiSCurrent staging systems for pancreatic cancerCancer J20121853949https://doi.org/10.1097/PPO.0b013e318278c5b5[PubMed][Google Scholar]
  • 33. FusaroliPKypraiosDCalettiGEloubeidiMAPancreatico-biliary endoscopic ultrasound: a systematic review of the levels of evidence, performance and outcomesWorld J Gastroenterol201218424356https://doi.org/10.3748/wjg.v18.i32.4243[PubMed][Google Scholar]
  • 34. ConradCFernandez-Del CastilloCPreoperative evaluation and management of the pancreatic head massJ Surg Oncol20131072332https://doi.org/10.1002/jso.23165[PubMed][Google Scholar]
  • 35. LeeESLeeJMImaging diagnosis of pancreatic cancer: A state-of-the-art reviewWorld J Gastroenterol201420786477https://doi.org/10.3748/wjg.v20.i24.7864[PubMed][Google Scholar]
  • 36. Mohammad AlizadehAHShahrokhSHadizadehMPadashiMZaliMRDiagnostic potency of EUS-guided FNA for the evaluation of pancreatic mass lesionsEndosc Ultrasound20165304https://doi.org/10.4103/2303-9027.175879[PubMed][Google Scholar]
  • 37. ShulikOCavanaghYGrossmanMPancreatic Lesion: Malignancy or Abscess?Am J Case Rep2016173379[PubMed][Google Scholar]
  • 38. PuliSRBechtoldMLBuxbaumJLEloubeidiMAHow good is endoscopic ultrasound-guided fine-needle aspiration in diagnosing the correct etiology for a solid pancreatic mass?: A meta-analysis and systematic reviewPancreas201342206https://doi.org/10.1097/MPA.0b013e3182546e79[PubMed][Google Scholar]
  • 39. DucreuxMCuhnaASCaramellaCHollebecqueABurtinPGoéréDSeufferleinTHaustermansKVan LaethemJLConroyTArnoldDCancer of the pancreas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-upAnnals of Oncology201526v56v68https://doi.org/10.1093/annonc/mdv295[PubMed][Google Scholar]
  • 40. RimbaşMDeaconuMCroitoruAHaidarASudden appearance of free fluid during endoscopic ultrasound-guided fine-needle aspirationEndosc Ultrasound20165557https://doi.org/10.4103/2303-9027.175900[PubMed][Google Scholar]
  • 41. FujiiLLLevyMJBasic techniques in endoscopic ultrasound-guided fine needle aspiration for solid lesions: Adverse events and avoiding themEndosc Ultrasound201433545https://doi.org/10.4103/2303-9027.123006[PubMed][Google Scholar]
  • 42. ChertoffJKhullarVBurkeLDuodenal perforation following esophagogastroduodenoscopy (EGD) with cautery and epinephrine injection for peptic ulcer disease: An interesting case of nonoperative management in the medical intensive care unit (MICU)Int J Surg Case Rep2015101215https://doi.org/10.1016/j.ijscr.2015.03.011[PubMed][Google Scholar]
  • 43. BournetBVignolle-VidoniAGrandDRoquesCBreibachFCrosJMuscariFCarrèreNSelvesJCordelierPBuscailLEndoscopic ultrasound-guided fine-needle aspiration plus KRAS and GNAS mutation in malignant intraductal papillary mucinous neoplasm of the pancreasEndosc Int Open20164E122835https://doi.org/10.1055/s-0042-117216[PubMed][Google Scholar]
  • 44. JonesSZhangXParsonsDWLinJCLearyRJAngenendtPMankooPCarterHKamiyamaHJimenoAHongSMFuBLinMTCore signaling pathways in human pancreatic cancers revealed by global genomic analysesScience200832118016https://doi.org/10.1126/science.1164368[PubMed][Google Scholar]
  • 45. HahnSAKernSEMolecular genetics of exocrine pancreatic neoplasmsSurg Clin North Am19957585769[PubMed][Google Scholar]
  • 46. KarhuRMahlamakiEKallioniemiAPancreatic adenocarcinoma — genetic portrait from chromosomes to microarraysGenes Chromosomes Cancer20064572130https://doi.org/10.1002/gcc.20337[PubMed][Google Scholar]
  • 47. CalhounESHuclTGallmeierEWestKMArkingDEMaitraAIacobuzio-DonahueCAChakravartiAHrubanRHKernSEIdentifying allelic loss and homozygous deletions in pancreatic cancer without matched normals using high-density single-nucleotide polymorphism arraysCancer Res20066679208https://doi.org/10.1158/0008-5472.can-06-0721[PubMed][Google Scholar]
  • 48. HaradaTChelalaCBhaktaVChaplinTCauleeKBarilPYoungBDLemoineNRGenome-wide DNA copy number analysis in pancreatic cancer using high-density single nucleotide polymorphism arraysOncogene200827195160https://doi.org/10.1038/sj.onc.1210832[PubMed][Google Scholar]
  • 49. BeroukhimRLinMParkYHaoKZhaoXGarrawayLAFoxEAHochbergEPMellinghoffIKHoferMDDescazeaudARubinMAMeyersonMInferring loss-of-heterozygosity from unpaired tumors using high-density oligonucleotide SNP arraysPLoS Comput Biol20062e41https://doi.org/10.1371/journal.pcbi.0020041[PubMed][Google Scholar]
  • 50. GreavesMMaleyCCClonal evolution in cancerNature201248130613http://www.nature.com/nature/journal/v481/n7381/abs/nature10762.html#supplementary-information[PubMed][Google Scholar]
  • 51. Makohon-MooreAIacobuzio-DonahueCAPancreatic cancer biology and genetics from an evolutionary perspectiveNat Rev Cancer20161655365https://doi.org/10.1038/nrc.2016.66[PubMed][Google Scholar]
  • 52. BardeesyNDePinhoRAPancreatic cancer biology and geneticsNat Rev Cancer20022897909https://doi.org/10.1038/nrc949[PubMed][Google Scholar]
  • 53. ChantrillLANagrialAMWatsonCJohnsALMartyn-SmithMSimpsonSMeadSJonesMDSamraJSGillAJWatsonNChinVTHumphrisJLPrecision Medicine for Advanced Pancreas Cancer: The Individualized Molecular Pancreatic Cancer Therapy (IMPaCT) TrialClin Cancer Res201521202937https://doi.org/10.1158/1078-0432.ccr-15-0426[PubMed][Google Scholar]
  • 54. YuJLiuSHSanchezRNemunaitisJRozengurtEBrunicardiFCPancreatic cancer actionable genes in precision medicine and personalized surgerySurgeon201715249https://doi.org/10.1016/j.surge.2016.05.002[PubMed][Google Scholar]
  • 55. NarayananVWeekesCDMolecular therapeutics in pancreas cancerWorld J Gastrointest Oncol2016836679https://doi.org/10.4251/wjgo.v8.i4.366[PubMed][Google Scholar]
  • 56. WongKQianZLeYThe Role of Precision Medicine in Pancreatic Cancer: Challenges for Targeted Therapy, Immune Modulating Treatment, Early Detection, and Less Invasive OperationsCancer Translational Medicine20162417https://doi.org/10.4103/2395-3977.181434[Google Scholar]
  • 57. San LucasFAAllensonKBernardVCastilloJKimDUEllisKEhliEADaviesGEPetersenJLLiDWolffRKatzMVaradhacharyGMinimally invasive genomic and transcriptomic profiling of visceral cancers by next-generation sequencing of circulating exosomesAnn Oncol20162763541https://doi.org/10.1093/annonc/mdv604[PubMed][Google Scholar]
  • 58. DiazLAJrBardelliALiquid biopsies: genotyping circulating tumor DNAJ Clin Oncol20143257986https://doi.org/10.1200/JCO.2012.45.2011[PubMed][Google Scholar]
  • 59. SiravegnaGBardelliABlood circulating tumor DNA for non-invasive genotyping of colon cancer patientsMol Oncol20161047580https://doi.org/10.1016/j.molonc.2015.12.005[PubMed][Google Scholar]
  • 60. RahbariMRahbariNReissfelderCWeitzJKahlertCExosomes: novel implications in diagnosis and treatment of gastrointestinal cancerLangenbecks Arch Surg20164011097110https://doi.org/10.1007/s00423-016-1468-2[PubMed][Google Scholar]
  • 61. ChengFSuLQianCCirculating tumor DNA: a promising biomarker in the liquid biopsy of cancerOncotarget201674883241https://doi.org/10.18632/oncotarget.9453[PubMed][Google Scholar]
  • 62. ThieleJABethelKKralickovaMKuhnPCirculating Tumor Cells: Fluid Surrogates of Solid TumorsAnnu Rev Pathol20171241947https://doi.org/10.1146/annurev-pathol-052016-100256[PubMed][Google Scholar]
  • 63. ShigeyasuKTodenSZumwaltTJOkugawaYGoelAEmerging Role of MicroRNAs as Liquid Biopsy Biomarkers in Gastrointestinal CancersClin Cancer Res20172323912399https://doi.org/10.1158/1078-0432.ccr-16-1676[PubMed][Google Scholar]
  • 64. VendrellJAMau-ThemFTBegantonBGodreuilSCoopmanPSolassolJCirculating Cell Free Tumor DNA Detection as a Routine Tool forLung Cancer Patient ManagementInt J Mol Sci201718https://doi.org/10.3390/ijms18020264[Google Scholar]
  • 65. LianidouESMavroudisDSotiropoulouGAgelakiSPantelKWhat's new on circulating tumor cells? A meeting reportBreast Cancer Res201012307https://doi.org/10.1186/bcr2601[PubMed][Google Scholar]
  • 66. LimSHBeckerTMChuaWCaixeiroNJNgWLKienzleNTognelaALumbaSRaskoJEde SouzaPSpringKJCirculating tumour cells and circulating free nucleic acid as prognostic and predictive biomarkers in colorectal cancerCancer Lett20143462433https://doi.org/10.1016/j.canlet.2013.12.019[PubMed][Google Scholar]
  • 67. VoelkerRLiquid biopsy receives approvalJAMA2016316260https://doi.org/10.1001/jama.2016.8833[Google Scholar]
  • 68. cobas EGFR Mutation Test v2Available online2016
  • 69. GuptaGPMassagueJCancer metastasis: building a frameworkCell200612767995https://doi.org/10.1016/j.cell.2006.11.001[PubMed][Google Scholar]
  • 70. RiquetMRiveraCGibaultLPricopiCMordantPBadiaAArameALe Pimpec BarthesF[Lymphatic spread of lung cancer: anatomical lymph node chains unchained in zones]. [Article in French]Rev Pneumol Clin2014701625https://doi.org/10.1016/j.pneumo.2013.07.001[PubMed][Google Scholar]
  • 71. PantelKSpeicherMRThe biology of circulating tumor cellsOncogene201635121624https://doi.org/10.1038/onc.2015.192[PubMed][Google Scholar]
  • 72. PantelKBrakenhoffRHDissecting the metastatic cascadeNat Rev Cancer2004444856https://doi.org/10.1038/nrc1370[PubMed][Google Scholar]
  • 73. KsiążkiewiczMMarkiewiczAŻaczek AJ. Epithelial-Mesenchymal Transition: A Hallmark in Metastasis Formation Linking Circulating Tumor Cells and Cancer Stem CellsPathobiology201279195208[PubMed][Google Scholar]
  • 74. WangRChadalavadaKWilshireJKowalikUHovingaKEGeberAFligelmanBLevershaMBrennanCTabarVGlioblastoma stem-like cells give rise to tumour endotheliumNature201046882933https://doi.org/10.1038/nature09624[PubMed][Google Scholar]
  • 75. Ricci-VitianiLPalliniRBiffoniMTodaroMInverniciGCenciTMairaGParatiEAStassiGLaroccaLMDe MariaRTumour vascularization via endothelial differentiation of glioblastoma stem-like cellsNature20104688248https://doi.org/10.1038/nature09557[PubMed][Google Scholar]
  • 76. KatsnelsonATumours grow their own blood vesselsNature News2010https://doi.org/doi:10.1038/news.2010.623[Google Scholar]
  • 77. FolbergRHendrixMJManiotisAJVasculogenic mimicry and tumor angiogenesisAm J Pathol200015636181https://doi.org/10.1016/s0002-9440(10)64739-6[PubMed][Google Scholar]
  • 78. FolbergRManiotisAJVasculogenic mimicryAPMIS200411250825https://doi.org/10.1111/j.1600-0463.2004.apm11207-0810.x[PubMed][Google Scholar]
  • 79. McDougallSRAndersonARChaplainMAMathematical modelling of dynamic adaptive tumour-induced angiogenesis: clinical implications and therapeutic targeting strategiesJ Theor Biol200624156489https://doi.org/10.1016/j.jtbi.2005.12.022[PubMed][Google Scholar]
  • 80. SpillFGuerreroPAlarconTMainiPKByrneHMMesoscopic and continuum modelling of angiogenesisJournal of Mathematical Biology201570485532https://doi.org/10.1007/s00285-014-0771-1[PubMed][Google Scholar]
  • 81. NozawaHChiuCHanahanDInfiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesisProc Natl Acad Sci U S A2006103124938https://doi.org/10.1073/pnas.0601807103[PubMed][Google Scholar]
  • 82. InoueMHagerJHFerraraNGerberHPHanahanDVEGF-A has a critical, nonredundant role in angiogenic switching and pancreatic beta cell carcinogenesisCancer Cell20021193202[PubMed][Google Scholar]
  • 83. BergersGBrekkenRMcMahonGVuTHItohTTamakiKTanzawaKThorpePItoharaSWerbZHanahanDMatrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesisNat Cell Biol2000273744https://doi.org/10.1038/35036374[PubMed][Google Scholar]
  • 84. AllardWJMateraJMillerMCRepolletMConnellyMCRaoCTibbeAGUhrJWTerstappenLWTumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseasesClin Cancer Res2004106897904https://doi.org/10.1158/1078-0432.ccr-04-0378[PubMed][Google Scholar]
  • 85. CristofanilliMBroglioKRGuarneriVJacksonSFritscheHAIslamRDawoodSReubenJMKauSWLaraJMKrishnamurthySUenoNTHortobagyiGNCirculating tumor cells in metastatic breast cancer: biologic staging beyond tumor burdenClin Breast Cancer200774719[PubMed][Google Scholar]
  • 86. RossJSSlodkowskaEACirculating and disseminated tumor cells in the management of breast cancerAm J Clin Pathol200913223745https://doi.org/10.1309/ajcpji7deolkcs6f[PubMed][Google Scholar]
  • 87. SzotekPPPieretti-VanmarckeRMasiakosPTDinulescuDMConnollyDFosterRDombkowskiDPrefferFMaclaughlinDTDonahoePKOvarian cancer side population defines cells with stem cell-like characteristics and Mullerian Inhibiting Substance responsivenessProc Natl Acad Sci U S A2006103111549https://doi.org/10.1073/pnas.0603672103[PubMed][Google Scholar]
  • 88. HaberDAVelculescuVEBlood-based analyses of cancer: circulating tumor cells and circulating tumor DNACancer Discov2014465061https://doi.org/10.1158/2159-8290.CD-13-1014[PubMed][Google Scholar]
  • 89. MasudaTHayashiNIguchiTItoSEguchiHMimoriKClinical and biological significance of circulating tumor cells in cancerMol Oncol20161040817https://doi.org/10.1016/j.molonc.2016.01.010[PubMed][Google Scholar]
  • 90. BraunSNaumeBCirculating and Disseminated Tumor CellsJournal of Clinical Oncology20052316236https://doi.org/doi:10.1200/JCO.2005.10.073[PubMed][Google Scholar]
  • 91. MohmeMRiethdorfSPantelKCirculating and disseminated tumour cells - mechanisms of immune surveillance and escapeNat Rev Clin Oncol201614155167https://doi.org/10.1038/nrclinonc.2016.144[PubMed][Google Scholar]
  • 92. FidlerIJThe pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisitedNat Rev Cancer200334538https://doi.org/10.1038/nrc1098[PubMed][Google Scholar]
  • 93. MassagueJObenaufACMetastatic colonization by circulating tumour cellsNature2016529298306https://doi.org/10.1038/nature17038[PubMed][Google Scholar]
  • 94. GiulianoMGiordanoAJacksonSDe GiorgiUMegoMCohenENGaoHAnfossiSHandyBCUenoNTAlvarezRHDe PlacidoSValeroVCirculating tumor cells as early predictors of metastatic spread in breast cancer patients with limited metastatic disseminationBreast Cancer Research201416440https://doi.org/10.1186/s13058-014-0440-8[PubMed][Google Scholar]
  • 95. XueJXieVKWangPCuiJGaoYLuZInterrelationships of Circulating Tumor Cells with Metastasis and Thrombosis: Role of MicroRNAsCurr Pharm Des2014205298308[PubMed][Google Scholar]
  • 96. FabisiewiczAGrzybowskaECTC clusters in cancer progression and metastasisMed Oncol20173412https://doi.org/10.1007/s12032-016-0875-0[PubMed][Google Scholar]
  • 97. MartinOAAndersonRLNarayanKMacManusMPDoes the mobilization of circulating tumour cells during cancer therapy cause metastasis?Nat Rev Clin Oncol2017143244https://doi.org/10.1038/nrclinonc.2016.128[PubMed][Google Scholar]
  • 98. KimMYOskarssonTAcharyyaSNguyenDXZhangXHFNortonLMassaguéJTumor Self-Seeding by Circulating Cancer CellsCell2009139131526https://doi.org/10.1016/j.cell.2009.11.025[PubMed][Google Scholar]
  • 99. ZheXCherMLBonfilRDCirculating tumor cells: finding the needle in the haystackAm J Cancer Res2011174051[PubMed][Google Scholar]
  • 100. SergeantGRoskamsTvan PeltJHoutmeyersFAertsRTopalBPerioperative cancer cell dissemination detected with a real-time RT-PCR assay for EpCAM is not associated with worse prognosis in pancreatic ductal adenocarcinomaBMC Cancer20111147https://doi.org/10.1186/1471-2407-11-47[PubMed][Google Scholar]
  • 101. DittamoreRLouwJKrupaRAnandADanilaDCArslanZBalesNMarrinucciDScherHIMolecular characterization of circulating tumor cells (CTC) and CTC subpopulations in progressive metastatic castration resistant prostate cancer (mCRPC)Journal of Clinical Oncology201432132https://doi.org/doi:10.1200/jco.2014.32.4_suppl.132[Google Scholar]
  • 102. PunnooseEAFerraldeschiRSzafer-GlusmanETuckerEKMohanSFlohrPRiisnaesRMirandaSFigueiredoIRodriguesDNOmlinAPezaroCZhuJPTEN loss in circulating tumour cells correlates with PTEN loss in fresh tumour tissue from castration-resistant prostate cancer patientsBr J Cancer2015113122533https://doi.org/10.1038/bjc.2015.332[PubMed][Google Scholar]
  • 103. HartIR‘Seed and soil’ revisited: mechanisms of site-specific metastasisCancer Metastasis Rev19821516https://doi.org/10.1007/bf00049477[PubMed][Google Scholar]
  • 104. ScottJKuhnPAndersonARUnifying metastasis--integrating intravasation, circulation and end-organ colonizationNat Rev Cancer2012124456https://doi.org/10.1038/nrc3287[PubMed][Google Scholar]
  • 105. QianCNTehBT“Seed and Soil” Theory of MetastasisSchwabMEncyclopedia of CancerBerlin, HeidelbergSpringer Berlin Heidelberg201133545
  • 106. HouJMKrebsMWardTSloaneRPriestLHughesAClackGRansonMBlackhallFDiveCCirculating tumor cells as a window on metastasis biology in lung cancerAm J Pathol201117898996https://doi.org/10.1016/j.ajpath.2010.12.003[PubMed][Google Scholar]
  • 107. MatsusakaSChinKOguraMSuenagaMShinozakiEMishimaYTeruiYMizunumaNHatakeKCirculating tumor cells as a surrogate marker for determining response to chemotherapy in patients with advanced gastric cancerCancer Sci2010101106771https://doi.org/10.1111/j.1349-7006.2010.01492.x[PubMed][Google Scholar]
  • 108. CohenSJPuntCJIannottiNSaidmanBHSabbathKDGabrailNYPicusJMorseMAMitchellEMillerMCDoyleGVTissingHTerstappenLWPrognostic significance of circulating tumor cells in patients with metastatic colorectal cancerAnn Oncol20092012239https://doi.org/10.1093/annonc/mdn786[PubMed][Google Scholar]
  • 109. Moussavi-HaramiSFWisinskiKBBeebeDJCirculating Tumor Cells in Metastatic Breast Cancer: A Prognostic and Predictive MarkerJ Patient Cent Res Rev201418592https://doi.org/10.17294/2330-0698.1017[PubMed][Google Scholar]
  • 110. HarrisLFritscheHMennelRNortonLRavdinPTaubeSSomerfieldMRHayesDFBastRCJrAmerican Society of Clinical Oncology2007update of recommendations for the use of tumor markers in breast cancerJ Clin Oncol2007255287312https://doi.org/10.1200/jco.2007.14.2364[Google Scholar]
  • 111. DanovaMTorchioMMazziniGIsolation of rare circulating tumor cells in cancer patients: technical aspects and clinical implicationsExpert Rev Mol Diagn20111147385https://doi.org/10.1586/erm.11.33[PubMed][Google Scholar]
  • 112. NagrathSSequistLVMaheswaranSBellDWIrimiaDUlkusLSmithMRKwakELDigumarthySMuzikanskyARyanPBalisUJTompkinsRGIsolation of rare circulating tumour cells in cancer patients by microchip technologyNature200745012359https://doi.org/10.1038/nature06385[PubMed][Google Scholar]
  • 113. BarriereGFiciPGalleraniGFabbriFZoliWRigaudMCirculating tumor cells and epithelial, mesenchymal and stemness markers: characterization of cell subpopulationsAnn Transl Med20142https://doi.org/10.3978/j.issn.2305-5839.2014.10.04[Google Scholar]
  • 114. MengSTripathyDFrenkelEPSheteSNaftalisEZHuthJFBeitschPDLeitchMHooverSEuhusDHaleyBMorrisonLFlemingTPCirculating tumor cells in patients with breast cancer dormancyClin Cancer Res200410815262https://doi.org/10.1158/1078-0432.ccr-04-1110[PubMed][Google Scholar]
  • 115. SollierEGoDECheJGossettDRO’ByrneSWeaverWMKummerNRettigMGoldmanJNickolsNMcCloskeySKulkarniRPDi CarloDSize-selective collection of circulating tumor cells using Vortex technologyLab Chip2014146377https://doi.org/10.1039/c3lc50689d[PubMed][Google Scholar]
  • 116. QinJAltJRHunsleyBAWilliamsTLFernandoMRStabilization of circulating tumor cells in blood using a collection device with a preservative reagentCancer Cell Int20141423https://doi.org/10.1186/1475-2867-14-23[PubMed][Google Scholar]
  • 117. TossAMuZFernandezSCristofanilliMCTC enumeration and characterization: moving toward personalized medicineAnn Transl Med20142https://doi.org/10.3978/j.issn.2305-5839.2014.09.06[Google Scholar]
  • 118. VonaGSabileALouhaMSitrukVRomanaSSchutzeKCapronFFrancoDPazzagliMVekemansMLacourBBrechotCPaterlini-BrechotPIsolation by size of epithelial tumor cells : a new method for the immunomorphological and molecular characterization of circulatingtumor cellsAm J Pathol20001565763https://doi.org/10.1016/s0002-9440(10)64706-2[PubMed][Google Scholar]
  • 119. HofmanVLongEIlieMBonnetaudCVignaudJMFlejouJFLantuejoulSPiatonEMouradNButoriCSelvaEMarquetteCHPoudenxMMorphological analysis of circulating tumour cells in patients undergoing surgery for non-small cell lung carcinoma using the isolation by size of epithelial tumour cell (ISET) methodCytopathology201223308https://doi.org/10.1111/j.1365-2303.2010.00835.x[PubMed][Google Scholar]
  • 120. HofmanVJIlieMIBonnetaudCSelvaELongEMolinaTVignaudJMFlejouJFLantuejoulSPiatonEButoriCMouradNPoudenxMCytopathologic detection of circulating tumor cells using the isolation by size of epithelial tumor cell method: promises and pitfallsAm J Clin Pathol201113514656https://doi.org/10.1309/ajcp9×8ozbeiqvvi[PubMed][Google Scholar]
  • 121. ZhangDZhaoLZhouPMaHHuangFJinMDaiXZhengXHuangSZhangTCirculating tumor microemboli (CTM) and vimentin+ circulating tumor cells (CTCs) detected by a size-based platform predict worse prognosis in advanced colorectal cancer patients during chemotherapyCancer Cell International2017176https://doi.org/10.1186/s12935-016-0373-7[PubMed][Google Scholar]
  • 122. KanwarNHuPBedardPClemonsMMcCreadyDDoneSJIdentification of genomic signatures in circulating tumor cells from breast cancerInternational Journal of Cancer201513733244https://doi.org/10.1002/ijc.29399[PubMed][Google Scholar]
  • 123. KalinichMBhanIKwanTTMiyamotoDTJavaidSLiCausiJAMilnerJDHongXGoyalLSilSChozMHoUKapurRAn RNA-based signature enables high specificity detection of circulating tumor cells in hepatocellular carcinomaProceedings of the National Academy of Sciences201711411238https://doi.org/10.1073/pnas.1617032114
  • 124. MarrinucciDBethelKKolatkarALuttgenMSMalchiodiMBaehringFVoigtKLazarDNievaJBazhenovaLKoAHKornWMSchramEFluid biopsy in patients with metastatic prostate, pancreatic and breast cancersPhys Biol20129016003https://doi.org/10.1088/1478-3975/9/1/016003[PubMed][Google Scholar]
  • 125. SchneckHGierkeBUppenkampFBehrensBNiederacherDStoeckleinNHTemplinMFPawlakMFehmTNeubauerHDisseminated Cancer Cell Network (DCC Net) DuesseldorfEpCAM-Independent Enrichment of Circulating Tumor Cells in Metastatic Breast CancerPLoS One201510e0144535https://doi.org/10.1371/journal.pone.0144535[PubMed][Google Scholar]
  • 126. SpiliotakiMMavroudisDKapranouKMarkomanolakiHKallergiGKoinisFKalbakisKGeorgouliasVAgelakiSEvaluation of proliferation and apoptosis markers in circulating tumor cells of women with early breast cancer who are candidates for tumor dormancyBreast Cancer Res201416https://doi.org/10.1186/s13058-014-0485-8[Google Scholar]
  • 127. BidardFCProudhonCPiergaJYCirculating tumor cells in breast cancerMolecular Oncology20161041830https://doi.org/http://dx.doi.org/10.1016/j.molonc.2016.01.001[PubMed][Google Scholar]
  • 128. LinHKZhengSWilliamsAJBalicMGroshenSScherHIFleisherMStadlerWDatarRHTaiYCCoteRJPortable filter-based microdevice for detection and characterization of circulating tumor cellsClin Cancer Res20101650118https://doi.org/10.1158/1078-0432.ccr-10-1105[PubMed][Google Scholar]
  • 129. Paterlini-BrechotPBenaliNLCirculating tumor cells (CTC) detection: clinical impact and future directionsCancer Lett2007253180204https://doi.org/10.1016/j.canlet.2006.12.014[PubMed][Google Scholar]
  • 130. FerraldeschiRMcDanielAKrupaRLouwJTuckerEBalesNMarrinucciDRiisnaesRMateoJDittamoreRBonoJSDTomlinsSAAttardGCK- and small nuclear size circulating tumor cell (CTCs) phenotypes in metastatic castration-resistant prostate cancer (mCRPC)Journal of Clinical Oncology201432209https://doi.org/10.1200/jco.2014.32.4_suppl.209[Google Scholar]
  • 131. AcetoNCirculating Tumor Cell Clusters are Oligoclonal Precursors of Breast Cancer MetastasisCell2014158111022https://doi.org/10.1016/j.cell.2014.07.013[PubMed][Google Scholar]
  • 132. HongYLiZZhangQA circulating tumor cell cluster-based model for tumor metastasis (Hypothesis)Oncol Lett20161248915https://doi.org/:10.3892/ol.2016.5358[PubMed][Google Scholar]
  • 133. ChoiJWKimJKYangYJKimPYoonKHYunSHUrokinase Exerts Antimetastatic Effects by Dissociating Clusters of Circulating Tumor CellsCancer Research201575447482https://doi.org/10.1158/0008-5472.can-15-0684[PubMed][Google Scholar]
  • 134. StottSLHsuCHTsukrovDIYuMMiyamotoDTWaltmanBARothenbergSMShahAMSmasMEKorirGKFloydFPJrGilmanAJLordJBIsolation of circulating tumor cells using a microvortex-generating herringbone-chipProc Natl Acad Sci U S A2010107183927https://doi.org/10.1073/pnas.1012539107[PubMed][Google Scholar]
  • 135. RiahiRGogoiPSepehriSZhouYHandiqueKGodseyJWangYA novel microchannel-based device to capture and analyze circulating tumor cells (CTCs) of breast cancerInt J Oncol20144418708https://doi.org/10.3892/ijo.2014.2353[PubMed][Google Scholar]
  • 136. BeijeNJagerASleijferSCirculating tumor cell enumeration by the CellSearch system: the clinician's guide to breast cancer treatment?Cancer Treat Rev20154114450https://doi.org/10.1016/j.ctrv.2014.12.008[PubMed][Google Scholar]
  • 137. BanerjeeSSKhobragadeVKhandareJDesigning Multicomponent Nanosystems for Rapid Detection of Circulating Tumor CellsMethods Mol Biol2017153027181https://doi.org/10.1007/978-1-4939-6646-2_16[PubMed][Google Scholar]
  • 138. ShengYWangTLiHZhangZChenJHeCLiYLvYZhangJXuCWangZHuangCWangLComparison of analytic performances of Cellsearch and iFISH approach in detecting circulating tumor cellsOncotarget2017888016https://doi.org/10.18632/oncotarget.6688[PubMed][Google Scholar]
  • 139. MingYLiYXingHLuoMLiZChenJMoJShiSCirculating Tumor Cells: From Theory to Nanotechnology-Based DetectionFront Pharmacol2017835https://doi.org/10.3389/fphar.2017.00035[PubMed][Google Scholar]
  • 140. WentPTLugliAMeierSBundiMMirlacherMSauterGDirnhoferSFrequent EpCam protein expression in human carcinomasHum Pathol2004351228[PubMed][Google Scholar]
  • 141. HyunKAJungHIAdvances and critical concerns with the microfluidic enrichments of circulating tumor cellsLab Chip2014144556https://doi.org/10.1039/c3lc50582k[PubMed][Google Scholar]
  • 142. LauniereCGaskillMCzaplewskiGMyungJHHongSEddingtonDTChannel surface patterning of alternating biomimetic protein combinations for enhanced microfluidic tumor cell isolationAnal Chem20128440228https://doi.org/10.1021/ac2033408[PubMed][Google Scholar]
  • 143. YoonHJKimTHZhangZAziziEPhamTMPaolettiCLinJRamnathNWichaMSHayesDFSimeoneDMNagrathSSensitive capture of circulating tumour cells by functionalized graphene oxide nanosheetsNat Nanotechnol2013873541https://doi.org/10.1038/nnano.2013.194[PubMed][Google Scholar]
  • 144. LiNXiaoTZhangZHeRWenDCaoYZhangWChenYA 3D graphene oxide microchip and a Au-enwrapped silica nanocomposite-based supersandwich cytosensor toward capture and analysis of circulating tumor cellsNanoscale201571635460https://doi.org/10.1039/c5nr04798f[PubMed][Google Scholar]
  • 145. MurlidharVZeinaliMGrabauskieneSGhannad-RezaieMWichaMSSimeoneDMRamnathNReddyRMNagrathSA radial flow microfluidic device for ultra-high-throughput affinity-based isolation of circulating tumor cellsSmall2014104895904https://doi.org/10.1002/smll.201400719[PubMed][Google Scholar]
  • 146. YoonHJShankerAWangYKozminskyMJinQPalanisamyNBurnessMLAziziESimeoneDMWichaMSKimJNagrathSTunable Thermal-Sensitive Polymer–Graphene Oxide Composite for Efficient Capture and Release of Viable Circulating Tumor CellsAdvanced Materials20162848917https://doi.org/10.1002/adma.201600658[PubMed][Google Scholar]
  • 147. ChenHCaoBSunBCaoYYangKLinYSChenHHighly-sensitive capture of circulating tumor cells using micro-ellipse filtersSci Rep20177610https://doi.org/10.1038/s41598-017-00232-6[PubMed][Google Scholar]
  • 148. KuskeAGorgesTMTennstedtPTiebelAKPompeRPreißerFPruesSMazelMMarkouALianidouEPeineSAlix-PanabièresCRiethdorfSImproved detection of circulating tumor cells in non-metastatic high-risk prostate cancer patientsScientific Reports2016639736https://doi.org/10.1038/srep39736[PubMed][Google Scholar]
  • 149. MüllerVRiethdorfSRackBJanniWFaschingPASolomayerEAktasBKasimir-BauerSPantelKFehmTPrognostic impact of circulating tumor cells assessed with the CellSearch System™ and AdnaTest Breast™ in metastatic breast cancer patients: the DETECT studyBreast Cancer Research201214R118https://doi.org/10.1186/bcr3243[PubMed][Google Scholar]
  • 150. SatelliABrownleeZMitraAMengQHLiSCirculating Tumor Cell Enumeration with a Combination of Epithelial Cell Adhesion Molecule– and Cell-Surface Vimentin–Based Methods for Monitoring Breast Cancer Therapeutic ResponseClinical Chemistry201461259[PubMed][Google Scholar]
  • 151. GroverPKCumminsAGPriceTJRoberts-ThomsonICHardinghamJECirculating tumour cells: the evolving concept and the inadequacy of their enrichment by EpCAM-based methodology for basic and clinical cancer researchAnnals of Oncology201425150616https://doi.org/10.1093/annonc/mdu018[PubMed][Google Scholar]
  • 152. WuSLiuSLiuZHuangJPuXLiJYangDDengHYangNXuJClassification of Circulating Tumor Cells by Epithelial-Mesenchymal Transition MarkersPLoS One201510e0123976https://doi.org/10.1371/journal.pone.0123976[PubMed][Google Scholar]
  • 153. KrawczykNMeier-StiegenFBanysMNeubauerHRuckhaeberleEFehmTExpression of Stem Cell and Epithelial-Mesenchymal Transition Markers in Circulating Tumor Cells of Breast Cancer PatientsBiomed Res Int20142014415721https://doi.org/10.1155/2014/415721[PubMed][Google Scholar]
  • 154. GorgesTMTinhoferIDroschMRoseLZollnerTMKrahnTvon AhsenOCirculating tumour cells escape from EpCAM-based detection due to epithelial-to-mesenchymal transitionBMC Cancer201212178https://doi.org/10.1186/1471-2407-12-178[PubMed][Google Scholar]
  • 155. PunnooseEAAtwalSKSpoerkeJMSavageHPanditaAYehRFPirzkallAFineBMAmlerLCChenDSLacknerMRMolecular biomarker analyses using circulating tumor cellsPLoS One20105e12517https://doi.org/10.1371/journal.pone.0012517[PubMed][Google Scholar]
  • 156. KonigsbergRObermayrEBisesGPfeilerGGneistMWrbaFde SantisMZeillingerRHudecMDittrichCDetection of EpCAM positive and negative circulating tumor cells in metastatic breast cancer patientsActa Oncol20115070010https://doi.org/10.3109/0284186x.2010.549151[PubMed][Google Scholar]
  • 157. YuMBardiaAWittnerBSStottSLSmasMETingDTIsakoffSJCicilianoJCWellsMNShahAMConcannonKFDonaldsonMCSequistLVCirculating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal compositionScience20133395804https://doi.org/10.1126/science.1228522[PubMed][Google Scholar]
  • 158. LiuQLiaoQZhaoYMyeloid-derived suppressor cells (MDSC) facilitate distant metastasis of malignancies by shielding circulating tumor cells (CTC) from immune surveillanceMed Hypotheses201687349https://doi.org/10.1016/j.mehy.2015.12.007[PubMed][Google Scholar]
  • 159. BalicMLinHWilliamsADatarRHCoteRJProgress in circulating tumor cell capture and analysis: implications for cancer managementExpert Review of Molecular Diagnostics20121230312https://doi.org/10.1586/erm.12.12[PubMed][Google Scholar]
  • 160. JueckstockJRackBFriedlTWScholzCSteidlJTrappETeschHForstbauerHLorenzRRezaiMHäberleLAlunni-FabbroniMSchneeweissADetection of circulating tumor cells using manually performed immunocytochemistry (MICC) does not correlate with outcome in patients with early breast cancer – Results of the German SUCCESS-A- trialBMC Cancer201616401https://doi.org/10.1186/s12885-016-2454-3[PubMed][Google Scholar]
  • 161. LowesLEAllanALRecent Advances in the Molecular Characterization of Circulating Tumor CellsCancers20146595624https://doi.org/10.3390/cancers6010595[PubMed][Google Scholar]
  • 162. OxnardGRPaweletzCPKuangYMachSLO’ConnellAMessineoMMLukeJJButaneyMKirschmeierPJackmanDMJännePANoninvasive detection of response and resistance in EGFR-mutant lung cancer using quantitative next-generation genotyping of cell-free plasma DNAClin Cancer Res2014201698705https://doi.org/10.1158/1078-0432.CCR-13-2482[PubMed][Google Scholar]
  • 163. PfitznerCSchröderIScheungraberCDoganARunnebaumIBDürstMHäfnerNDigital-Direct-RT-PCR: a sensitive and specific method for quantification of CTC in patients with cervical carcinomaScientific Reports201443970https://doi.org/10.1038/srep03970https://www.nature.com/articles/srep03970#supplementary-information[PubMed][Google Scholar]
  • 164. DiamondELeeGYAkhtarNHKirbyBJGiannakakouPTagawaSTNanusDMIsolation and characterization of circulating tumor cells in prostate cancerFrontiers in Oncology20122131https://doi.org/10.3389/fonc.2012.00131[PubMed][Google Scholar]
  • 165. DenisJAPatroniAGuillermEPepinDBenali-FuretNWechslerJManceauGBernardMCouletFLarsenAKKarouiMLacorteJMDroplet digital PCR of circulating tumor cells from colorectal cancer patients can predict KRAS mutations before surgeryMol Oncol201610122131https://doi.org/10.1016/j.molonc.2016.05.009[PubMed][Google Scholar]
  • 166. FreidinMBFreydinaDVLeungMMontero FernandezANicholsonAGLimECirculating tumor DNA outperforms circulating tumor cells for KRAS mutation detection in thoracic malignanciesClin Chem2015611299304https://doi.org/10.1373/clinchem.2015.242453[PubMed][Google Scholar]
  • 167. Castellanos-RizaldosEMilburyCAGuhaMMakrigiorgosGMCOLD-PCR enriches low-level variant DNA sequences and increases the sensitivity of genetic testingMethods Mol Biol2014110262339https://doi.org/10.1007/978-1-62703-727-3_33[PubMed][Google Scholar]
  • 168. YeeSSLiebermanDBBlanchardTRaderJZhaoJTroxelABDeSlooverDFoxAJDaberRDKakrechaBSukhadiaSBelkaGKDeMicheleAMA novel approach for next-generation sequencing of circulating tumor cellsMol Genet Genomic Med20164395406https://doi.org/10.1002/mgg3.210[PubMed][Google Scholar]
  • 169. PerakisSSpeicherMREmerging concepts in liquid biopsiesBMC Medicine20171575https://doi.org/10.1186/s12916-017-0840-6[PubMed][Google Scholar]
  • 170. LohrJGAdalsteinssonVACibulskisKChoudhuryADRosenbergMCruz-GordilloPFrancisJMZhangCZShalekAKSatijaRTrombettaJJLuDTallapragadaNWhole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancerNat Biotechnol20143247984https://doi.org/10.1038/nbt.2892[PubMed][Google Scholar]
  • 171. HindsonBJNessKDMasquelierDABelgraderPHerediaNJMakarewiczAJBrightIJLuceroMYHiddessenALLeglerTCKitanoTKHodelMRPetersenJFHigh-throughput droplet digital PCR system for absolute quantitation of DNA copy numberAnal Chem201183860410https://doi.org/10.1021/ac202028g[PubMed][Google Scholar]
  • 172. Tellez-GabrielMOryBLamoureuxFHeymannMFHeymannDTumour Heterogeneity: The Key Advantages of Single-Cell AnalysisInternational Journal of Molecular Sciences2016172142https://doi.org/10.3390/ijms17122142[Google Scholar]
  • 173. BiWStambrookPJDetection of known mutation by proof-reading PCRNucleic Acids Research19982630735[PubMed][Google Scholar]
  • 174. BarnardRFutoVPecheniukNSlatteryMWalshTPCR bias toward the wild-type k-ras and p53 sequences: implications for PCR detection of mutations and cancer diagnosisBiotechniques19982568491[PubMed][Google Scholar]
  • 175. LiJMakrigiorgosGMCOLD-PCR: a new platform for highly improved mutation detection in cancer and genetic testingBiochem Soc Trans20093742732https://doi.org/10.1042/bst0370427[PubMed][Google Scholar]
  • 176. BabayanAAlawiMGormleyMMullerVWikmanHMcMullinRPSmirnovDALiWGeffkenMPantelKJoosseSAComparative study of whole genome amplification and next generation sequencing performance of single cancer cellsOncotarget201685606656080https://doi.org/10.18632/oncotarget.10701[PubMed][Google Scholar]
  • 177. El MessaoudiSRoletFMouliereFThierryARCirculating cell free DNA: Preanalytical considerationsClin Chim Acta201342422230https://doi.org/10.1016/j.cca.2013.05.022[PubMed][Google Scholar]
  • 178. MalentacchiFPizzamiglioSVerderioPPazzagliMOrlandoCCiniselliCMGuntherKGelminiSInfluence of storage conditions and extraction methods on the quantity and quality of circulating cell-free DNA (ccfDNA): the SPIDIA-DNAplas External Quality Assessment experienceClin Chem Lab Med201553193542https://doi.org/10.1515/cclm-2014-1161[PubMed][Google Scholar]
  • 179. PantelKBrakenhoffRHBrandtBDetection, clinical relevance and specific biological properties of disseminating tumour cellsNat Rev Cancer2008832940https://doi.org/10.1038/nrc2375[PubMed][Google Scholar]
  • 180. Iwanicki-CaronIBasilePToureEAntoniettiMLecleireSDi FioreAOden-GangloffABlanchardFLemoineFDi FioreFSabourinJCMichelPUsefulness of circulating tumor cell detection in pancreatic adenocarcinoma diagnosisAm J Gastroenterol20131081525https://doi.org/10.1038/ajg.2012.367[PubMed][Google Scholar]
  • 181. RhimADThegeFISantanaSMLanninTBSahaTNTsaiSMaggsLRKochmanMLGinsbergGGLiebJGChandrasekharaVDrebinJAAhmadNDetection of Circulating Pancreas Epithelial Cells in Patients with Pancreatic Cystic LesionsGastroenterology201414664751https://doi.org/10.1053/j.gastro.2013.12.007[PubMed][Google Scholar]
  • 182. EarlJGarcia-NietoSMartinez-AvilaJCMontansJSanjuanbenitoARodriguez-GarroteMLisaEMendiaELoboEMalatsNCarratoAGuillen-PonceCCirculating tumor cells (Ctc) and kras mutant circulating free Dna (cfdna) detection in peripheral blood as biomarkers in patients diagnosed with exocrine pancreatic cancerBMC Cancer201515797https://doi.org/10.1186/s12885-015-1779-7[PubMed][Google Scholar]
  • 183. ZhangYWangFNingNChenQYangZGuoYXuDZhangDZhanTCuiWPatterns of circulating tumor cells identified by CEP8, CK and CD45 in pancreatic cancerInt J Cancer2015136122833https://doi.org/10.1002/ijc.29070[PubMed][Google Scholar]
  • 184. BobekVGurlichREliasovaPKolostovaKCirculating tumor cells in pancreatic cancer patients: Enrichment and cultivationWorld J Gastroenterol2014201716370https://doi.org/10.3748/wjg.v20.i45.17163[PubMed][Google Scholar]
  • 185. KulemannBLissASWarshawALSeifertSBronsertPGlatzTPitmanMBHoeppnerJKRAS mutations in pancreatic circulating tumor cells: a pilot studyTumour Biol201637754754https://doi.org/10.1007/s13277-015-4589-2[PubMed][Google Scholar]
  • 186. GaoYZhuYZhangZZhangCHuangXYuanZClinical significance of pancreatic circulating tumor cells using combined negative enrichment and immunostaining-fluorescence in situ hybridizationJ Exp Clin Cancer Res20163566https://doi.org/10.1186/s13046-016-0340-0[PubMed][Google Scholar]
  • 187. AnkenyJSCourtCMHouSLiQSongMWuDChenJFLeeTLinMShoSRochefortMMGirgisMDYaoJCirculating tumour cells as a biomarker for diagnosis and staging in pancreatic cancerBr J Cancer2016114136775https://doi.org/10.1038/bjc.2016.121[PubMed][Google Scholar]
  • 188. PorukKEBlackfordALWeissMJCameronJLHeJGogginsMRasheedZAWolfgangCLWoodLDCirculating Tumor Cells Expressing Markers of Tumor-Initiating Cells Predict Poor Survival and Cancer Recurrence in Patients with Pancreatic Ductal AdenocarcinomaClin Cancer Res20172326812690https://doi.org/10.1158/1078-0432.ccr-16-1467[PubMed][Google Scholar]
  • 189. ZhouJHuLYuZZhengJYangDBouvetMHoffmanRMMarker expression in circulating cancer cells of pancreatic cancer patientsJ Surg Res20111716316https://doi.org/10.1016/j.jss.2010.05.007[PubMed][Google Scholar]
  • 190. KhojaLBackenASloaneRMenasceLRyderDKrebsMBoardRClackGHughesABlackhallFValleJWDiveCA pilot study to explore circulating tumour cells in pancreatic cancer as a novel biomarkerBr J Cancer201210650816https://doi.org/10.1038/bjc.2011.545[PubMed][Google Scholar]
  • 191. HanLChenWZhaoQPrognostic value of circulating tumor cells in patients with pancreatic cancer: a meta-analysisTumour Biol201435247380https://doi.org/10.1007/s13277-013-1327-5[PubMed][Google Scholar]
  • 192. YuMTingDTStottSLWittnerBSOzsolakFPaulSCicilianoJCSmasMEWinokurDGilmanAJUlmanMJXegaKContinoGRNA sequencing of pancreatic circulating tumour cells implicates WNT signaling in metastasisNature20124875103https://doi.org/10.1038/nature11217[PubMed][Google Scholar]
  • 193. KomarGKauhanenSLiukkoKSeppanenMKajanderSOvaskaJNuutilaPMinnHDecreased blood flow with increased metabolic activity: a novel sign of pancreatic tumor aggressivenessClin Cancer Res20091555117https://doi.org/10.1158/1078-0432.ccr-09-0414[PubMed][Google Scholar]
  • 194. CatenacciDVTChapmanCGXuPKoonsAKondaVJSiddiquiUDWaxmanIAcquisition of Portal Venous Circulating Tumor Cells From Patients With Pancreaticobiliary Cancers by Endoscopic UltrasoundGastroenterology20151491794803.e4https://doi.org/10.1053/j.gastro.2015.08.050[PubMed][Google Scholar]
  • 195. YuMBardiaAAcetoNBersaniFMaddenMWDonaldsonMCDesaiRZhuHComaillsVZhengZWittnerBSStojanovPBrachtelECancer therapy. Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibilityScience201434521620https://doi.org/10.1126/science.1253533[PubMed][Google Scholar]
  • 196. BobekVKacprzakGRzechonekAKolostovaKDetection and cultivation of circulating tumor cells in malignant pleural mesotheliomaAnticancer Res20143425659[PubMed][Google Scholar]
  • 197. BobekVMatkowskiRGurlichRGrabowskiKSzelachowskaJLischkeRSchutznerJHarustiakTPazdroARzechonekAKolostovaKCultivation of circulating tumor cells in esophageal cancerFolia Histochem Cytobiol2014521717https://doi.org/10.5603/fhc.2014.0020[PubMed][Google Scholar]
  • 198. CeganMKolostovaKMatkowskiRBroulMSchramlJFiutowskiMBobekVIn vitro culturing of viable circulating tumor cells of urinary bladder cancerInt J Clin Exp Pathol20147716471[PubMed][Google Scholar]
  • 199. ZhangZShiratsuchiHLinJChenGReddyRMAziziEFouladdelSChangACLinLJiangHWaghrayMLukerGSimeoneDMExpansion of CTCs from early stage lung cancer patients using a microfluidic co-culture modelOncotarget201451238397https://doi.org/10.18632/oncotarget.2592[PubMed][Google Scholar]
  • 200. CayrefourcqLMazardTJoosseSSolassolJRamosJAssenatESchumacherUCostesVMaudelondeTPantelKAlix-PanabieresCEstablishment and characterization of a cell line from human circulating colon cancer cellsCancer Res201575892901https://doi.org/10.1158/0008-5472.can-14-2613[PubMed][Google Scholar]
  • 201. Voskoglou-NomikosTPaterJLSeymourLClinical predictive value of the in vitro cell line, human xenograft, and mouse allograft preclinical cancer modelsClin Cancer Res20039422739[PubMed][Google Scholar]
  • 202. JohnsonJIDeckerSZaharevitzDRubinsteinLVVendittiJMSchepartzSKalyandrugSChristianMArbuckSHollingsheadMSausvilleEARelationships between drug activity in NCI preclinical in vitro and in vivo models and early clinical trialsBr J Cancer200184142431https://doi.org/10.1054/bjoc.2001.1796[PubMed][Google Scholar]
  • 203. EndDWSmetsGToddAVApplegateTLFueryCJAngibaudPVenetMSanzGPoignetHSkrzatSDevineAWoutersWBowdenCCharacterization of the antitumor effects of the selective farnesyl protein transferase inhibitor R115777 in vivo and in vitroCancer Res2001611317[PubMed][Google Scholar]
  • 204. Van CutsemEvan de VeldeHKarasekPOettleHVervenneWLSzawlowskiASchoffskiPPostSVerslypeCNeumannHSafranHHumbletYPerez RuixoJPhase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancerJ Clin Oncol20042214308https://doi.org/10.1200/jco.2004.10.112[PubMed][Google Scholar]
  • 205. MerkleFTGhoshSKamitakiNMitchellJAviorYMelloCKashinSMekhoubadSIlicDCharltonMSaphierGHandsakerREGenoveseGHuman pluripotent stem cells recurrently acquire and expand dominant negative P53 mutationsNature201754522933https://doi.org/10.1038/nature22312[PubMed][Google Scholar]
  • 206. HodgkinsonCLMorrowCJLiYMetcalfRLRothwellDGTrapaniFPolanskiRBurtDJSimpsonKLMorrisKPepperSDNonakaDGreystokeATumorigenicity and genetic profiling of circulating tumor cells in small-cell lung cancerNat Med201420897903https://doi.org/10.1038/nm.3600[PubMed][Google Scholar]
  • 207. BaccelliISchneeweissARiethdorfSStenzingerASchillertAVogelVKleinCSainiMBauerleTWallwienerMHolland-LetzTHofnerTSprickMIdentification of a population of blood circulating tumor cells from breast cancer patients that initiates metastasis in a xenograft assayNat Biotechnol20133153944https://doi.org/10.1038/nbt.2576[PubMed][Google Scholar]
  • 208. RossiERuggeMFacchinettiAPizziMNardoGBarbieriVManiconeMDe FaveriSChiara ScainiMBassoUAmadoriAZamarchiRRetaining the long-survive capacity of Circulating Tumor Cells (CTCs) followed by xeno-transplantation: not only from metastatic cancer of the breast but also of prostate cancer patientsOncoscience201414956https://doi.org/10.18632/oncoscience.8[PubMed][Google Scholar]
  • 209. WilliamsESRodriguez-BravoVChippada-VenkataUDe Ia Iglesia-VicenteJGongYGalskyMOhWCordon-CardoCDomingo-DomenechJGeneration of Prostate Cancer Patient Derived Xenograft Models from Circulating Tumor CellsJ Vis Exp201553182https://doi.org/10.3791/53182[PubMed][Google Scholar]
  • 210. GaoDVelaISbonerAIaquintaPJKarthausWRGopalanADowlingCWanjalaJNUndvallEAAroraVKWongvipatJKossaiMRamazanogluSOrganoid cultures derived from patients with advanced prostate cancerCell201415917687https://doi.org/10.1016/j.cell.2014.08.016[PubMed][Google Scholar]
  • 211. GiulianoMHerreraSChristinyPShawCCreightonCJMitchellTBhatRZhangXMaoSDobroleckiLEAl-rawiAChenFVenezianiBMCirculating and disseminated tumor cells from breast cancer patient-derived xenograft-bearing mice as a novel model to study metastasisBreast Cancer Research2015173https://doi.org/10.1186/s13058-014-0508-5[PubMed][Google Scholar]
  • 212. KhooBLGrenciGLiquid biopsy and therapeutic response: Circulating tumor cell cultures for evaluation of anticancer treatmentSci Adv20162e1600274https://doi.org/10.1126/sciadv.1600274[PubMed][Google Scholar]
  • 213. TorphyRJTignanelliCJKamandeJWMoffittRAHerrera LoezaSGSoperSAYehJJCirculating tumor cells as a biomarker of response to treatment in patient-derived xenograft mouse models of pancreatic adenocarcinomaPLoS One20149e89474https://doi.org/10.1371/journal.pone.0089474[PubMed][Google Scholar]
  • 214. YuKHRiciglianoMHidalgoMAbou-AlfaGKLoweryMASaltzLBCrottyJFGaryKCooperBLapidusRSadowskaMO’ReillyEMPharmacogenomic Modeling of Circulating Tumor and Invasive Cells for Prediction of Chemotherapy Response and Resistance in Pancreatic CancerClinical Cancer Research20142052819https://doi.org/10.1158/1078-0432.ccr-14-0531[PubMed][Google Scholar]
  • 215. MansourHCell-free nucleic acids as noninvasive biomarkers for colorectal cancer detectionFront Genet20145https://doi.org/10.3389/fgene.2014.00182[PubMed][Google Scholar]
  • 216. DiehlFSchmidtKChotiMARomansKGoodmanSLiMThorntonKAgrawalNSokollLSzaboSAKinzlerKWVogelsteinBDiazLAJrCirculating mutant DNA to assess tumor dynamicsNat Med20081498590https://doi.org/10.1038/nm.1789[PubMed][Google Scholar]
  • 217. KohlerCBarekatiZRadpourRZhongXYCell-free DNA in the circulation as a potential cancer biomarkerAnticancer Res20113126238[PubMed][Google Scholar]
  • 218. SchwarzenbachHNishidaNCalinGAPantelKClinical relevance of circulating cell-free microRNAs in cancerNat Rev Clin Oncol20141114556https://doi.org/10.1038/nrclinonc.2014.5[PubMed][Google Scholar]
  • 219. AkcaHDemirayAYarenABirFKoselerAIwakawaRBagciGYokotaJUtility of serum DNA and pyrosequencing for the detection of EGFR mutations in non-small cell lung cancerCancer Genet20132067380https://doi.org/10.1016/j.cancergen.2013.01.005[PubMed][Google Scholar]
  • 220. StrounMAnkerPNucleic acids spontaneously released by living frog auriclesBiochem J1972128100p1p[Google Scholar]
  • 221. StrounMLyauteyJLederreyCOlson-SandAAnkerPAbout the possible origin and mechanism of circulating DNA apoptosis and active DNA releaseClin Chim Acta200131313942[PubMed][Google Scholar]
  • 222. RogersJCBoldtDKornfeldSSkinnerAValeriCRExcretion of deoxyribonucleic acid by lymphocytes stimulated with phytohemagglutinin or antigenProc Natl Acad Sci U S A19726916859[PubMed][Google Scholar]
  • 223. AnkerPStrounMMauricePASpontaneous release of DNA by human blood lymphocytes as shown in an in vitro systemCancer Res197535237582[PubMed][Google Scholar]
  • 224. DelgadoPOAlvesBCGehrke FdeSKuniyoshiRKWroclavskiMLDel GiglioAFonsecaFLCharacterization of cell-free circulating DNA in plasma in patients with prostate cancerTumour Biol2013349836https://doi.org/10.1007/s13277-012-0634-6[PubMed][Google Scholar]
  • 225. RoninsonIBBroudeEVChangBDIf not apoptosis, then what? Treatment-induced senescence and mitotic catastrophe in tumor cellsDrug Resist Updat2001430313https://doi.org/10.1054/drup.2001.0213[PubMed][Google Scholar]
  • 226. LaktionovPPTamkovichSNRykovaEYBryzgunovaOEStarikovAVKuznetsovaNPSumarokovSVKolomietsSASevostianovaNVVlassovVVExtracellular circulating nucleic acids in human plasma in health and diseaseNucleosides Nucleotides Nucleic Acids20042387983https://doi.org/10.1081/ncn-200026035[PubMed][Google Scholar]
  • 227. LoYMChanLYLoKWLeungSFZhangJChanATLeeJCHjelmNMJohnsonPJHuangDPQuantitative analysis of cell-free Epstein-Barr virus DNA in plasma of patients with nasopharyngeal carcinomaCancer Res199959118891[PubMed][Google Scholar]
  • 228. YangHJLiuVWTsangPCYipAMTamKFWongLCNgTYNganHYQuantification of human papillomavirus DNA in the plasma of patients with cervical cancerInt J Gynecol Cancer20041490310https://doi.org/10.1111/j.1048-891X.2004.014528.x[PubMed][Google Scholar]
  • 229. OnoAFujimotoAYamamotoYAkamatsuSHiragaNImamuraMKawaokaTTsugeMAbeHHayesCNMikiDFurutaMTsunodaTCirculating Tumor DNA Analysis for Liver Cancers and Its Usefulness as a Liquid BiopsyCell Mol Gastroenterol Hepatol2015151634https://doi.org/10.1016/j.jcmgh.2015.06.009[PubMed][Google Scholar]
  • 230. HuangXO’ConnorRKwizeraEAGold Nanoparticle Based Platforms for Circulating Cancer Marker DetectionNanotheranostics2017180102https://doi.org/10.7150/ntno.18216[PubMed][Google Scholar]
  • 231. TamkovichSNCherepanovaAVKolesnikovaEVRykovaEYPyshnyiDVVlassovVVLaktionovPPCirculating DNA and DNase activity in human bloodAnn N Y Acad Sci200610751916https://doi.org/10.1196/annals.1368.026[PubMed][Google Scholar]
  • 232. TsuiNBJiangPChowKCSuXLeungTYSunHChanKCChiuRWLoYMHigh resolution size analysis of fetal DNA in the urine of pregnant women by paired-end massively parallel sequencingPLoS One20127e48319https://doi.org/10.1371/journal.pone.0048319[PubMed][Google Scholar]
  • 233. ReckampKLMelnikovaVOKarlovichCSequistLVCamidgeDRWakeleeHPerolMOxnardGRKoscoKCroucherPSamuelszEVibatCRGuerreroSA Highly Sensitive and Quantitative Test Platform for Detection of NSCLC EGFR Mutations in Urine and PlasmaJ Thorac Oncol2016111690700https://doi.org/10.1016/j.jtho.2016.05.035[PubMed][Google Scholar]
  • 234. ChanKCJiangPChanCWSunKWongJHuiEPChanSLChanWCHuiDSNgSSChanHLWongCSMaBBNoninvasive detection of cancer-associated genome-wide hypomethylation and copy number aberrations by plasma DNA bisulfite sequencingProc Natl Acad Sci U S A2013110187618https://doi.org/10.1073/pnas.1313995110[PubMed][Google Scholar]
  • 235. ChusedTMSteinbergADTalalNThe clearance and localization of nucleic acids by New Zealand and normal miceClinical and Experimental Immunology19721246576[PubMed][Google Scholar]
  • 236. HeitzerEAuerMHoffmannEMPichlerMGaschCUlzPLaxSWaldispuehl-GeiglJMauermannOMohanSPristauzGLacknerCHöflerGEstablishment of tumor-specific copy number alterations from plasma DNA of patients with cancerInt J Cancer201313334656https://doi.org/10.1002/ijc.28030[PubMed][Google Scholar]
  • 237. ThierryARMouliereFGongoraCOllierJRobertBYchouMDel RioMMolinaFOrigin and quantification of circulating DNA in mice with human colorectal cancer xenograftsNucleic Acids Res201038615975https://doi.org/10.1093/nar/gkq421[PubMed][Google Scholar]
  • 238. MouliereFRobertBArnau PeyrotteEDel RioMYchouMMolinaFGongoraCThierryARHigh fragmentation characterizes tumour-derived circulating DNAPLoS One20116e23418https://doi.org/10.1371/journal.pone.0023418[PubMed][Google Scholar]
  • 239. ChanKCZhangJHuiABWongNLauTKLeungTNLoKWHuangDWLoYMSize distributions of maternal and fetal DNA in maternal plasmaClin Chem2004508892https://doi.org/10.1373/clinchem.2003.024893[PubMed][Google Scholar]
  • 240. ToEWChanKCLeungSFChanLYToKFChanATJohnsonPJLoYMRapid clearance of plasma Epstein-Barr virus DNA after surgical treatment of nasopharyngeal carcinomaClin Cancer Res2003932549[PubMed][Google Scholar]
  • 241. YaoWMeiCNanXHuiLEvaluation and comparison of in vitro degradation kinetics of DNA in serum, urine and saliva: A qualitative studyGene20165901428https://doi.org/10.1016/j.gene.2016.06.033[PubMed][Google Scholar]
  • 242. JiangPLoYMDThe Long and Short of Circulating Cell-Free DNA and the Ins and Outs of Molecular DiagnosticsTrends Genet20163236071https://doi.org/10.1016/j.tig.2016.03.009[PubMed][Google Scholar]
  • 243. GalluzziLKroemerGSecondary Necrosis: Accidental No MoreTrends Cancer2017312https://doi.org/10.1016/j.trecan.2016.12.001[PubMed][Google Scholar]
  • 244. WangBGHuangHYChenYCBristowREKassaueiKChengCCRodenRSokollLJChanDWShih IeMIncreased plasma DNA integrity in cancer patientsCancer Res20036339668[PubMed][Google Scholar]
  • 245. JahrSHentzeHEnglischSHardtDFackelmayerFOHeschRDKnippersRDNA Fragments in the Blood Plasma of Cancer Patients: Quantitations and Evidence for Their Origin from Apoptotic and Necrotic CellsCancer Research200161165965[PubMed][Google Scholar]
  • 246. RaymondCKHernandezJKarrRHillKLiMCollection of cell-free DNA for genomic analysis of solid tumors in a clinical laboratory settingPLoS One201712e0176241https://doi.org/10.1371/journal.pone.0176241[PubMed][Google Scholar]
  • 247. JiangWWZahurakMGoldenbergDMilmanYParkHLWestraWHKochWSidranskyDCalifanoJIncreased plasma DNA integrity index in head and neck cancer patientsInt J Cancer200611926736https://doi.org/10.1002/ijc.22250[PubMed][Google Scholar]
  • 248. ZaherERAnwarMMKohailHMAEl-ZoghbySMAbo-El-EneenMSValue of circulating DNA concentration and integrity as a screening test for detection of cancer in an Egyptian cohortAlexandria Journal of Medicine20124818796https://doi.org/http://dx.doi.org/10.1016/j.ajme.2012.03.003[Google Scholar]
  • 249. KamelAMTeamaSFawzyAEl DeftarMPlasma DNA integrity index as a potential molecular diagnostic marker for breast cancerTumour Biol201637756572https://doi.org/10.1007/s13277-015-4624-3[PubMed][Google Scholar]
  • 250. UnderhillHRKitzmanJOHellwigSWelkerNCDazaRBakerDNGligorichKMRostomilyRCBronnerMPShendureJFragment Length of Circulating Tumor DNAPLoS Genetics201612e1006162https://doi.org/10.1371/journal.pgen.1006162[PubMed][Google Scholar]
  • 251. SunKJiangPChanKCWongJChengYKLiangRHChanWKMaESChanSLChengSHChanRWTongYKNgSSPlasma DNA tissue mapping by genome-wide methylation sequencing for noninvasive prenatal, cancer, and transplantation assessmentsProc Natl Acad Sci U S A2015112E550312https://doi.org/10.1073/pnas.1508736112[PubMed][Google Scholar]
  • 252. StrounMMauricePVasioukhinVLyauteyJLederreyCLefortFRossierAChenXQAnkerPThe origin and mechanism of circulating DNAAnn N Y Acad Sci20009061618[PubMed][Google Scholar]
  • 253. StrounMLyauteyJLederreyCMulcahyHEAnkerPAlu repeat sequences are present in increased proportions compared to a unique gene in plasma/serum DNA: evidence for a preferential release from viable cells?Ann N Y Acad Sci200194525864[PubMed][Google Scholar]
  • 254. RogersJCIdentification of an intracellular precursor to DNA excreted by human lymphocytesProc Natl Acad Sci U S A19767332115[PubMed][Google Scholar]
  • 255. KostiukSVMalinovskaiaEMErmakovAVSmirnovaTDKamenevaLVChvartatskaiaOVLosevaPAErshovaESLiubchenkoLNVeikoNN[Cell-free DNA fragments increase transcription in human mesenchymal stem cells, activate TLR-dependent signal pathway and supress apoptosis]. [Article in Russian]Biomed Khim20125867383[PubMed][Google Scholar]
  • 256. FleischhackerMSchmidtBCirculating nucleic acids (CNAs) and cancer—a surveyBiochim Biophys Acta20071775181232https://doi.org/10.1016/j.bbcan.2006.10.001[PubMed][Google Scholar]
  • 257. ElshimaliYIKhaddourHSarkissyanMWuYVadgamaJVThe Clinical Utilization of Circulating Cell Free DNA (CCFDNA) in Blood of Cancer PatientsInt J Mol Sci2013141892558https://doi.org/10.3390/ijms140918925[PubMed][Google Scholar]
  • 258. MarzeseDMHiroseHHoonDSDiagnostic and prognostic value of circulating tumor-related DNA in cancer patientsExpert Rev Mol Diagn20131382744https://doi.org/10.1586/14737159.2013.845088[PubMed][Google Scholar]
  • 259. BettegowdaCSausenMLearyRJKindeIWangYAgrawalNBartlettBRWangHLuberBAlaniRMAntonarakisESAzadNSBardelliADetection of Circulating Tumor DNA in Early- and Late-Stage Human MalignanciesSci Transl Med20146224ra24https://doi.org/10.1126/scitranslmed.3007094[Google Scholar]
  • 260. SiravegnaGMarsoniSSienaSBardelliAIntegrating liquid biopsies into the management of cancerNat Rev Clin Oncol201714531548https://doi.org/10.1038/nrclinonc.2017.14[PubMed][Google Scholar]
  • 261. HeitzerEUlzPGeiglJBCirculating tumor DNA as a liquid biopsy for cancerClin Chem20156111223https://doi.org/10.1373/clinchem.2014.222679[PubMed][Google Scholar]
  • 262. QinZLjubimovVAZhouCTongYLiangJCell-free circulating tumor DNA in cancerChin J Cancer20163536https://doi.org/10.1186/s40880-016-0092-4[PubMed][Google Scholar]
  • 263. BeckJOellerichMSchulzUSchauerteVReinhardLFuchsUKnabbeCZittermannAOlbrichtCGummertJFShipkovaMBirschmannIWielandEDonor-Derived Cell-Free DNA Is a Novel Universal Biomarker for Allograft Rejection in Solid Organ TransplantationTransplant Proc20154724003https://doi.org/10.1016/j.transproceed.2015.08.035[PubMed][Google Scholar]
  • 264. BeckJBierauSBalzerSAndagRKanzowPSchmitzJGaedckeJMoererOSlottaJEWalsonPKollmarOOellerichMSchutzEDigital droplet PCR for rapid quantification of donor DNA in the circulation of transplant recipients as a potential universal biomarker of graft injuryClin Chem201359173241https://doi.org/10.1373/clinchem.2013.210328[PubMed][Google Scholar]
  • 265. ZouJDuffyBSladeMYoungALStewardNHachemRMohanakumarTRapid detection of donor cell free DNA in lung transplant recipients with rejections using donor-recipient HLA mismatchHum Immunol201778342349https://doi.org/10.1016/j.humimm.2017.03.002[PubMed][Google Scholar]
  • 266. AdamekMOpelzGKleinKMorathCTranTHA fast and simple method for detecting and quantifying donor-derived cell-free DNA in sera of solid organ transplant recipients as a biomarker for graft functionClin Chem Lab Med201654114755https://doi.org/10.1515/cclm-2015-0622[PubMed][Google Scholar]
  • 267. GrskovicMHillerDJEubankLASninskyJJChristophersonCCollinsJPThompsonKSongMWangYSRossDNellesMJYeeJPWilberJCValidation of a Clinical-Grade Assay to Measure Donor-Derived Cell-Free DNA in Solid Organ Transplant RecipientsJ Mol Diagn201618890902https://doi.org/10.1016/j.jmoldx.2016.07.003[PubMed][Google Scholar]
  • 268. BloomRDBrombergJSPoggioEDBunnapradistSLangoneAJSoodPMatasAJMehtaSMannonRBSharfuddinAFischbachBNarayananMJordanSCCell-Free DNA and Active Rejection in Kidney AllograftsJ Am Soc Nephrol20172822212232https://doi.org/10.1681/asn.2016091034[PubMed][Google Scholar]
  • 269. ShapiroBChakrabartyMCohnEMLeonSADetermination of circulating DNA levels in patients with benign or malignant gastrointestinal diseaseCancer198351211620[PubMed][Google Scholar]
  • 270. SilvaJMGarciaJMDominguezGSilvaJMirallesCCantosBCocaSProvencioMEspanaPBonillaFPersistence of tumor DNA in plasma of breast cancer patients after mastectomyAnn Surg Oncol20029716[PubMed][Google Scholar]
  • 271. SozziGConteDLeonMCiricioneRRozLRatcliffeCRozECireneiNBellomiMPelosiGPierottiMAPastorinoUQuantification of free circulating DNA as a diagnostic marker in lung cancerJ Clin Oncol20032139028https://doi.org/10.1200/jco.2003.02.006[PubMed][Google Scholar]
  • 272. RenNYeQHQinLXZhangBHLiuYKTangZYCirculating DNA level is negatively associated with the long-term survival of hepatocellular carcinoma patientsWorld J Gastroenterol20061239114[PubMed][Google Scholar]
  • 273. BoddyJLGalSMalonePRHarrisALWainscoatJSProspective study of quantitation of plasma DNA levels in the diagnosis of malignant versus benign prostate diseaseClin Cancer Res20051113949https://doi.org/10.1158/1078-0432.ccr-04-1237[PubMed][Google Scholar]
  • 274. BankiFMasonRJOhDHagenJADeMeesterSRLiphamJCTanakaKDanenbergKDYacoubWNDanenbergPVDeMeesterTRPlasma DNA as a molecular marker for completeness of resection and recurrent disease in patients with esophageal cancerArch Surg20071425338https://doi.org/10.1001/archsurg.142.6.533[PubMed][Google Scholar]
  • 275. DiazLAJrWilliamsRTWuJKindeIHechtJRBerlinJAllenBBozicIReiterJGNowakMAKinzlerKWOlinerKSVogelsteinBThe molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancersNature201248653740https://doi.org/10.1038/nature11219[PubMed][Google Scholar]
  • 276. ForshewTMurtazaMParkinsonCGaleDTsuiDWKaperFDawsonSJPiskorzAMJimenez-LinanMBentleyDHadfieldJMayAPCaldasCNoninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNASci Transl Med20124136ra68https://doi.org/10.1126/scitranslmed.3003726[Google Scholar]
  • 277. McBrideDJOrpanaAKSotiriouCJoensuuHStephensPJMudieLJHamalainenEStebbingsLAAnderssonLCFlanaganAMDurbecqVIgnatiadisMKallioniemiOUse of cancer-specific genomic rearrangements to quantify disease burden in plasma from patients with solid tumorsGenes Chromosomes Cancer20104910629https://doi.org/10.1002/gcc.20815[PubMed][Google Scholar]
  • 278. Garcia-OlmoDCSamosJPicazoMGAsensioAITobosoIGarcia-OlmoDRelease of cell-free DNA into the bloodstream leads to high levels of non-tumor plasma DNA during tumor progression in ratsCancer Lett200827213340https://doi.org/10.1016/j.canlet.2008.07.003[PubMed][Google Scholar]
  • 279. Gonzalez-MasiaJAGarcia-OlmoDGarcia-OlmoDCCirculating nucleic acids in plasma and serum (CNAPS): applications in oncologyOnco Targets Ther2013681932https://doi.org/10.2147/ott.s44668[PubMed][Google Scholar]
  • 280. MisaleSYaegerRHoborSScalaEJanakiramanMLiskaDValtortaESchiavoRBuscarinoMSiravegnaGBencardinoKCercekAChenCTEmergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancerNature20124865326https://doi.org/10.1038/nature11156[PubMed][Google Scholar]
  • 281. MurtazaMDawsonSJTsuiDWGaleDForshewTPiskorzAMParkinsonCChinSFKingsburyZWongASMarassFHumphraySHadfieldJNon-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNANature201349710812https://doi.org/10.1038/nature12065[PubMed][Google Scholar]
  • 282. SalviSGurioliGDe GiorgiUConteducaVTedaldiGCalistriDCasadioVCell-free DNA as a diagnostic marker for cancer: current insightsOnco Targets Ther20169654959https://doi.org/10.2147/ott.s100901[PubMed][Google Scholar]
  • 283. van 't VeerLJDaiHvan de VijverMJHeYDHartAAMaoMPeterseHLvan der KooyKMartonMJWitteveenATSchreiberGJKerkhovenRMRobertsCGene expression profiling predicts clinical outcome of breast cancerNature20024155306https://doi.org/10.1038/415530a[PubMed][Google Scholar]
  • 284. DawsonSJTsuiDWMurtazaMBiggsHRuedaOMChinSFDunningMJGaleDForshewTMahler-AraujoBRajanSHumphraySBecqJAnalysis of circulating tumor DNA to monitor metastatic breast cancerN Engl J Med20133681199209https://doi.org/10.1056/NEJMoa1213261[PubMed][Google Scholar]
  • 285. DiehlFLiMDressmanDHeYShenDSzaboSDiazLAJrGoodmanSNDavidKAJuhlHKinzlerKWVogelsteinBDetection and quantification of mutations in the plasma of patients with colorectal tumorsProc Natl Acad Sci U S A20051021636873https://doi.org/10.1073/pnas.0507904102[PubMed][Google Scholar]
  • 286. ChanKCJiangPZhengYWLiaoGJSunHWongJSiuSSChanWCChanSLChanATLaiPBChiuRWLoYMCancer genome scanning in plasma: detection of tumor-associated copy number aberrations, single-nucleotide variants, and tumoral heterogeneity by massively parallel sequencingClin Chem20135921124https://doi.org/10.1373/clinchem.2012.196014[PubMed][Google Scholar]
  • 287. BohersEViaillyPJDuboisSBertrandPMaingonnatCMareschalSRuminyPPicquenotJMBastardCDesmotsFFestTLeroyKTillyHSomatic mutations of cell-free circulating DNA detected by next-generation sequencing reflect the genetic changes in both germinal center B-cell-like and activated B-cell-like diffuse large B-cell lymphomas at the time of diagnosisHaematologica2015100e2804https://doi.org/10.3324/haematol.2015.123612[PubMed][Google Scholar]
  • 288. ParkinsonCAGaleDPiskorzAMBiggsHHodgkinCAddleyHFreemanSMoylePSalaESayalKHoskingKGounarisIJimenez-LinanMExploratory Analysis of TP53 Mutations in Circulating Tumour DNA as Biomarkers of Treatment Response for Patients with Relapsed High-Grade Serous Ovarian Carcinoma: A Retrospective StudyPLoS Medicine201613e1002198https://doi.org/10.1371/journal.pmed.1002198[PubMed][Google Scholar]
  • 289. TalyVPekinDBenhaimLKotsopoulosSKLe CorreDLiXAtochinILinkDRGriffithsADPallierKBlonsHBoucheOLandiBMultiplex picodroplet digital PCR to detect KRAS mutations in circulating DNA from the plasma of colorectal cancer patientsClin Chem201359172231https://doi.org/10.1373/clinchem.2013.206359[PubMed][Google Scholar]
  • 290. HuangAZhangXZhouSLCaoYHuangXWFanJYangXRZhouJDetecting Circulating Tumor DNA in Hepatocellular Carcinoma Patients Using Droplet Digital PCR Is Feasible and Reflects Intratumoral HeterogeneityJ Cancer20167190714https://doi.org/10.7150/jca.15823[PubMed][Google Scholar]
  • 291. DressmanDYanHTraversoGKinzlerKWVogelsteinBTransforming single DNA molecules into fluorescent magnetic particles for detection and enumeration of genetic variationsProc Natl Acad Sci U S A2003100881722https://doi.org/10.1073/pnas.1133470100[PubMed][Google Scholar]
  • 292. DiehlFLiMHeYKinzlerKWVogelsteinBDressmanDBEAMing: single-molecule PCR on microparticles in water-in-oil emulsionsNat Methods200635519https://doi.org/10.1038/nmeth898[PubMed][Google Scholar]
  • 293. NewmanAMBratmanSVToJWynneJFEclovNCModlinLALiuCLNealJWWakeleeHAMerrittREShragerJBLooBWJrAn ultrasensitive method for quantitating circulating tumor DNA with broad patient coverageNat Med20142054854https://doi.org/10.1038/nm.3519[PubMed][Google Scholar]
  • 294. KindeIWuJPapadopoulosNKinzlerKWVogelsteinBDetection and quantification of rare mutations with massively parallel sequencingProc Natl Acad Sci U S A201110895305https://doi.org/10.1073/pnas.1105422108[PubMed][Google Scholar]
  • 295. KennedySRDetecting ultralow-frequency mutations by Duplex SequencingNat Protoc201492586606https://doi.org/10.1038/nprot.2014.170[PubMed][Google Scholar]
  • 296. NewmanAMLovejoyAFKlassDMKurtzDMChabonJJSchererFStehrHLiuCLBratmanSVSayCZhouLCarterJNWestRBIntegrated digital error suppression for improved detection of circulating tumor DNANat Biotechnol20163454755https://doi.org/10.1038/nbt.3520[PubMed][Google Scholar]
  • 297. HeitzerEUlzPBelicJGutschiSQuehenbergerFFischerederKBenezederTAuerMPischlerCMannweilerSTumor-associated copy number changes in the circulation of patients with prostate cancer identified through whole-genome sequencingGenome Med20135https://doi.org/10.1186/gm434[PubMed][Google Scholar]
  • 298. LearyRJSausenMKindeIPapadopoulosNCarptenJDCraigDO’ShaughnessyJKinzlerKWParmigianiGVogelsteinBDetection of chromosomal alterations in the circulation of cancer patients with whole-genome sequencingSci Transl Med20124https://doi.org/10.1126/scitranslmed.3004742[Google Scholar]
  • 299. PietraszDPecuchetNGarlanFDidelotADubreuilODoatSImbert-BismutFKarouiMVaillantJCTalyVLaurent-PuigPBachetJBPlasma Circulating Tumor DNA in Pancreatic Cancer Patients Is a Prognostic MarkerClin Cancer Res20172311623https://doi.org/10.1158/1078-0432.ccr-16-0806[PubMed][Google Scholar]
  • 300. XueXTeareMDHolenIZhuYMWollPJOptimizing the yield and utility of circulating cell-free DNA from plasma and serumClin Chim Acta20094041004https://doi.org/10.1016/j.cca.2009.02.018[PubMed][Google Scholar]
  • 301. SherwoodJLCorcoranCBrownHSharpeADMusilovaMKohlmannAOptimised Pre-Analytical Methods Improve KRAS Mutation Detection in Circulating Tumour DNA (ctDNA) from Patients with Non-Small Cell Lung Cancer (NSCLC)PLoS One201611https://doi.org/10.1371/journal.pone.0150197[Google Scholar]
  • 302. ValleeAMarcqMBizieuxAKouriCELacroixHBennounaJDouillardJYDenisMGPlasma is a better source of tumor-derived circulating cell-free DNA than serum for the detection of EGFR alterations in lung tumor patientsLung Cancer2013823734https://doi.org/10.1016/j.lungcan.2013.08.014[PubMed][Google Scholar]
  • 303. MaMZhuHZhangCSunXGaoXChenG“Liquid biopsy”—ctDNA detection with great potential and challengesAnn Transl Med20153https://doi.org/10.3978/j.issn.2305-5839.2015.09.29[Google Scholar]
  • 304. YiXMaJGuanYChenRYangLXiaXThe feasibility of using mutation detection in ctDNA to assess tumor dynamicsInt J Cancer201714026427https://doi.org/10.1002/ijc.30620[PubMed][Google Scholar]
  • 305. NgSBChuaCNgMGanAPoonPSTeoMFuCLeowWQLimKHChungAKooSLChooSPHoDIndividualised multiplexed circulating tumour DNA assays for monitoring of tumour presence in patients after colorectal cancer surgerySci Rep20177https://doi.org/10.1038/srep40737[PubMed][Google Scholar]
  • 306. MaireFMicardSHammelPVoitotHLévyPCugnencPRuszniewskiPPuigPLDifferential diagnosis between chronic pancreatitis and pancreatic cancer: value of the detection of KRAS2 mutations in circulating DNABr J Cancer2002875514https://doi.org/10.1038/sj.bjc.6600475[PubMed][Google Scholar]
  • 307. BergerAWSchwerdelDCostaIGHackertTStrobelOLamSBarthTFSchroppelBMeiningABuchlerMWZenkeMHermannPCSeufferleinTDetection of Hot-Spot Mutations in Circulating Cell-Free DNA From Patients With Intraductal Papillary Mucinous Neoplasms of the PancreasGastroenterology201615126770https://doi.org/10.1053/j.gastro.2016.04.034[PubMed][Google Scholar]
  • 308. ChengHLiuCJiangJLuoGLuYJinKGuoMZhangZXuJLiuLNiQYuXAnalysis of ctDNA to predict prognosis and monitor treatment responses in metastatic pancreatic cancer patientsInt J Cancer2017140234450https://doi.org/10.1002/ijc.30650[PubMed][Google Scholar]
  • 309. ZillOAGreeneCSebisanovicDSiewLMLengJVuMHendifarAEWangZAtreyaCEKelleyRKVan LoonKKoAHTemperoMACell-Free DNA Next-Generation Sequencing in Pancreatobiliary CarcinomasCancer Discov2015510408https://doi.org/10.1158/2159-8290.cd-15-0274[PubMed][Google Scholar]
  • 310. SausenMPhallenJAdleffVJonesSLearyRJBarrettMTAnagnostouVParpart-LiSMurphyDKay LiQHrubanCAScharpfRWhiteJRClinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patientsNat Commun201567686https://doi.org/10.1038/ncomms8686[PubMed][Google Scholar]
  • 311. HenriksenSDMadsenPHLarsenACJohansenMBDrewesAMPedersenISKrarupHThorlacius-UssingOCell-free DNA promoter hypermethylation in plasma as a diagnostic marker for pancreatic adenocarcinomaClin Epigenetics20168117https://doi.org/10.1186/s13148-016-0286-2[PubMed][Google Scholar]
  • 312. TjensvollKLapinMBuhlTOltedalSSteen-Ottosen BerryKGiljeBSoreideJAJavleMNordgardOSmaalandRClinical relevance of circulating KRAS mutated DNA in plasma from patients with advanced pancreatic cancerMol Oncol20161063543https://doi.org/10.1016/j.molonc.2015.11.012[PubMed][Google Scholar]
  • 313. SinghNGuptaSPandeyRMChauhanSSSarayaAHigh levels of cell-free circulating nucleic acids in pancreatic cancer are associated with vascular encasement, metastasis and poor survivalCancer Invest2015337885https://doi.org/10.3109/07357907.2014.1001894[PubMed][Google Scholar]
  • 314. BryantKLManciasJDKimmelmanACDerCJKRAS: feeding pancreatic cancer proliferationTrends Biochem Sci20143991100https://doi.org/10.1016/j.tibs.2013.12.004[PubMed][Google Scholar]
  • 315. EserSSchniekeASchneiderGSaurDOncogenic KRAS signalling in pancreatic cancerBr J Cancer201411181722https://doi.org/10.1038/bjc.2014.215[PubMed][Google Scholar]
  • 316. KandaMMatthaeiHWuJHongSMYuJBorgesMHrubanRHMaitraAKinzlerKVogelsteinBGogginsMPresence of somatic mutations in most early-stage pancreatic intraepithelial neoplasiaGastroenterology20121427303.e9https://doi.org/10.1053/j.gastro.2011.12.042[PubMed][Google Scholar]
  • 317. YachidaSWhiteCMNaitoYZhongYBrosnanJAMacgregor-DasAMMorganRASaundersTLaheruDAHermanJMHrubanRHKleinAPJonesSClinical Significance of the Genetic Landscape of Pancreatic Cancer and Implications for Identification of Potential Long Term SurvivorsClin Cancer Res201218633947https://doi.org/10.1158/1078-0432.CCR-12-1215[PubMed][Google Scholar]
  • 318. WitkiewiczAKMcMillanEABalajiUBaekGHLinWCMansourJMollaeeMWagnerKUKoduruPYoppAChotiMAYeoCJMcCuePWhole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targetsNat Commun201566744https://doi.org/10.1038/ncomms7744[PubMed][Google Scholar]
  • 319. KinugasaHNousoKMiyaharaKMorimotoYDohiCTsutsumiKKatoHMatsubaraTOkadaHYamamotoKDetection of K-ras gene mutation by liquid biopsy in patients with pancreatic cancerCancer2015121227180https://doi.org/10.1002/cncr.29364[PubMed][Google Scholar]
  • 320. MatsubayashiHCantoMSatoNKleinAAbeTYamashitaKYeoCJKallooAHrubanRGogginsMDNA methylation alterations in the pancreatic juice of patients with suspected pancreatic diseaseCancer Res200666120817https://doi.org/10.1158/0008-5472.can-05-2664[PubMed][Google Scholar]
  • 321. LiggettTMelnikovAYiQLReplogleCBrandRKaulKTalamontiMAbramsRALevensonVDifferential methylation of cell-free circulating DNA among patients with pancreatic cancer versus chronic pancreatitisCancer2010116167480https://doi.org/10.1002/cncr.24893[PubMed][Google Scholar]
  • 322. Nogueira da CostaAHercegZDetection of cancer-specific epigenomic changes in biofluids: powerful tools in biomarker discovery and applicationMol Oncol2012670415https://doi.org/10.1016/j.molonc.2012.07.005[PubMed][Google Scholar]
  • 323. SatoNGogginsMThe role of epigenetic alterations in pancreatic cancerJ Hepatobiliary Pancreat Surg20061328695https://doi.org/10.1007/s00534-005-1057-1[PubMed][Google Scholar]
  • 324. TanACJimenoALinSHWheelhouseJChanFSolomonARajeshkumarNVRubio-ViqueiraBHidalgoMCharacterizing DNA methylation patterns in pancreatic cancer genomeMolecular Oncology2009342538https://doi.org/10.1016/j.molonc.2009.03.004[PubMed][Google Scholar]
  • 325. WartonKMahonKLSamimiGMethylated circulating tumor DNA in blood: power in cancer prognosis and responseEndocrine-Related Cancer201623R157R71https://doi.org/10.1530/ERC-15-0369[PubMed][Google Scholar]
  • 326. ThompsonMJRubbiLDawsonDWDonahueTRPellegriniMPancreatic Cancer Patient Survival Correlates with DNA Methylation of Pancreas Development GenesPLoS One201510e0128814https://doi.org/10.1371/journal.pone.0128814[PubMed][Google Scholar]
  • 327. McDonaldOGLiXSaundersTTryggvadottirRMentchSJWarmoesMOWordAECarrerASalzTHNatsumeSStaufferKMMakohon-MooreAZhongYEpigenomic reprogramming during pancreatic cancer progression links anabolic glucose metabolism to distant metastasisNat Genet20174936776https://doi.org/10.1038/ng.3753[PubMed][Google Scholar]
  • 328. Lehmann-WermanRNeimanDZemmourHMossJMagenheimJVaknin-DembinskyARubertssonSNellgårdBBlennowKZetterbergHSpaldingKHallerMJWasserfallCHIdentification of tissue-specific cell death using methylation patterns of circulating DNAProc Natl Acad Sci U S A2016113E1826E34https://doi.org/10.1073/pnas.1519286113[PubMed][Google Scholar]
  • 329. MoranSMartinez-CardusASayolsSMusulenEBalanaCEstival-GonzalezAMoutinhoCHeynHDiaz-LagaresAde MouraMCStellaGMComoglioPMRuiz-MiroMEpigenetic profiling to classify cancer of unknown primary: a multicentre, retrospective analysisLancet Oncol201617138695https://doi.org/10.1016/s1470-2045(16)30297-2[PubMed][Google Scholar]
  • 330. MarusykAPolyakKTumor heterogeneity: causes and consequencesBiochim Biophys Acta2010180510517https://doi.org/10.1016/j.bbcan.2009.11.002[PubMed][Google Scholar]
  • 331. BedardPLHansenARRatainMJSiuLLTumour heterogeneity in the clinicNature201350135564https://doi.org/10.1038/nature12627[PubMed][Google Scholar]
  • 332. MurtazaMDawsonSJPogrebniakKRuedaOMProvenzanoEGrantJChinSFTsuiDWMarassFGaleDAliHRShahPContente-CuomoTMultifocal clonal evolution characterized using circulating tumour DNA in a case of metastatic breast cancerNat Commun201568760https://doi.org/10.1038/ncomms9760[PubMed][Google Scholar]
  • 333. WillmsEJohanssonHJMägerILeeYBlombergKESadikMAlaargASmithCILehtiöJEl AndaloussiSWoodMJVaderPCells release subpopulations of exosomes with distinct molecular and biological propertiesScientific Reports2016622519https://doi.org/10.1038/srep22519[PubMed][Google Scholar]
  • 334. BoukourisSMathivananSExosomes in bodily fluids are a highly stable resource of disease biomarkersProteomics Clin Appl2015935867https://doi.org/10.1002/prca.201400114[PubMed][Google Scholar]
  • 335. JohnstoneRMAdamMHammondJROrrLTurbideCVesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes)J Biol Chem1987262941220[PubMed][Google Scholar]
  • 336. TheryCZitvogelLAmigorenaSExosomes: composition, biogenesis and functionNat Rev Immunol2002256979https://doi.org/10.1038/nri855[PubMed][Google Scholar]
  • 337. PanBTTengKWuCAdamMJohnstoneRMElectron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytesJ Cell Biol19851019428[PubMed][Google Scholar]
  • 338. ValadiHEkstromKBossiosASjostrandMLeeJJLotvallJOExosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cellsNat Cell Biol200796549https://doi.org/10.1038/ncb1596[PubMed][Google Scholar]
  • 339. AlexanderMHuRRuntschMCKageleDAMosbrugerTLTolmachovaTSeabraMCRoundJLWardDMO’ConnellRMExosome-delivered microRNAs modulate the inflammatory response to endotoxinNature Communications201567321https://doi.org/10.1038/ncomms8321[Google Scholar]
  • 340. TanCYLaiRCWongWDanYYLimSKHoHKMesenchymal stem cell-derived exosomes promote hepatic regeneration in drug-induced liver injury modelsStem Cell Research & Therapy2014576https://doi.org/10.1186/scrt465[PubMed][Google Scholar]
  • 341. KellerSSandersonMPStoeckAAltevogtPExosomes: from biogenesis and secretion to biological functionImmunol Lett20061071028https://doi.org/10.1016/j.imlet.2006.09.005[PubMed][Google Scholar]
  • 342. AdmyreCJohanssonSMQaziKRFilenJJLahesmaaRNormanMNeveEPScheyniusAGabrielssonSExosomes with immune modulatory features are present in human breast milkJ Immunol2007179196978[PubMed][Google Scholar]
  • 343. SimhadriVRReinersKSHansenHPTopolarDSimhadriVLNohroudiKKuferTAEngertAPogge von StrandmannEDendritic cells release HLA-B-associated transcript-3 positive exosomes to regulate natural killer functionPLoS One20083e3377https://doi.org/10.1371/journal.pone.0003377[PubMed][Google Scholar]
  • 344. BahriniISongJHDiezDHanayamaRNeuronal exosomes facilitate synaptic pruning by up-regulating complement factors in microgliaScientific Reports201557989https://doi.org/10.1038/srep07989[PubMed][Google Scholar]
  • 345. EngelmannBMassbergSThrombosis as an intravascular effector of innate immunityNat Rev Immunol2013133445https://doi.org/10.1038/nri3345[PubMed][Google Scholar]
  • 346. GuayCRegazziRCirculating microRNAs as novel biomarkers for diabetes mellitusNat Rev Endocrinol2013951321https://doi.org/10.1038/nrendo.2013.86[PubMed][Google Scholar]
  • 347. LeeMJParkDHKangJHExosomes as the source of biomarkers of metabolic diseasesAnn Pediatr Endocrinol Metab20162111925https://doi.org/10.6065/apem.2016.21.3.119[PubMed][Google Scholar]
  • 348. BoulangerCMLoyerXRautouPEAmabileNExtracellular vesicles in coronary artery diseaseNat Rev Cardiol20171425972https://doi.org/10.1038/nrcardio.2017.7[PubMed][Google Scholar]
  • 349. PerrottaIAquilaSExosomes in human atherosclerosis: An ultrastructural analysis studyUltrastruct Pathol2016401016https://doi.org/10.3109/01913123.2016.1154912[PubMed][Google Scholar]
  • 350. MasyukAIMasyukTVLarussoNFExosomes in the pathogenesis, diagnostics and therapeutics of liver diseasesJ Hepatol2013596215https://doi.org/10.1016/j.jhep.2013.03.028[PubMed][Google Scholar]
  • 351. VellaLJSharplesRANisbetRMCappaiRHillAFThe role of exosomes in the processing of proteins associated with neurodegenerative diseasesEur Biophys J20083732332https://doi.org/10.1007/s00249-007-0246-z[PubMed][Google Scholar]
  • 352. AzmiASBaoBSarkarFHExosomes in Cancer Development, Metastasis and Drug Resistance: A Comprehensive ReviewCancer Metastasis Rev20133210.1007/s10555-013-9441-9. https://doi.org/10.1007/s10555-013-9441-9[Google Scholar]
  • 353. HerringJMMcMichaelMASmithSAMicroparticles in health and diseaseJ Vet Intern Med201327102033https://doi.org/10.1111/jvim.12128[PubMed][Google Scholar]
  • 354. CiardielloCCavalliniLSpinelliCYangJReis-SobreiroMde CandiaPMinciacchiVRDi VizioDFocus on Extracellular Vesicles: New Frontiers of Cell-to-Cell Communication in CancerInt J Mol Sci201617175https://doi.org/10.3390/ijms17020175[PubMed][Google Scholar]
  • 355. KalluriRLeBleuVSDiscovery of Double-Stranded Genomic DNA in Circulating ExosomesCold Spring Harb Symp Quant Biol201681275280https://doi.org/10.1101/sqb.2016.81.030932[PubMed][Google Scholar]
  • 356. BeachAZhangHGRatajczakMZKakarSSExosomes: an overview of biogenesis, composition and role in ovarian cancerJournal of Ovarian Research2014714https://doi.org/10.1186/1757-2215-7-14[PubMed][Google Scholar]
  • 357. ThakurBKZhangHBeckerAMateiIHuangYCosta-SilvaBZhengYHoshinoABrazierHXiangJWilliamsCRodriguez-BarruecoRSilvaJMDouble-stranded DNA in exosomes: a novel biomarker in cancer detectionCell Res2014247669https://doi.org/10.1038/cr.2014.44[PubMed][Google Scholar]
  • 358. ThomasCEEhrhardtAKayMAProgress and problems with the use of viral vectors for gene therapyNat Rev Genet2003434658https://doi.org/10.1038/nrg1066[PubMed][Google Scholar]
  • 359. FreedmanJEGersteinMMickERozowskyJLevyDKitchenRDasSShahRDanielsonKBeaulieuLNavarroFCWangYGaleevTRDiverse human extracellular RNAs are widely detected in human plasmaNature Communications2016711106https://doi.org/10.1038/ncomms11106[Google Scholar]
  • 360. HuangXYuanTTschannenMSunZJacobHDuMLiangMDittmarRLLiuYLiangMKohliMThibodeauSNBoardmanLCharacterization of human plasma-derived exosomal RNAs by deep sequencingBMC Genomics201314319https://doi.org/10.1186/1471-2164-14-319[PubMed][Google Scholar]
  • 361. MathivananSFahnerCJReidGESimpsonRJExoCarta 2012: database of exosomal proteins, RNA and lipidsNucleic Acids Res201240D12414https://doi.org/10.1093/nar/gkr828[PubMed][Google Scholar]
  • 362. KeerthikumarSChisangaDAriyaratneDSaffarHAAnandSZhaoKSamuelMPathanMJoisMChilamkurtiNGangodaLMathivananSExoCarta: A web-based compendium of exosomal cargoJ Mol Biol201642868892https://doi.org/10.1016/j.jmb.2015.09.019[PubMed][Google Scholar]
  • 363. AdmyreCGrunewaldJThybergJGripenbackSTornlingGEklundAScheyniusAGabrielssonSExosomes with major histocompatibility complex class II and co-stimulatory molecules are present in human BAL fluidEur Respir J20032257883[PubMed][Google Scholar]
  • 364. ZhangHGGrizzleWEExosomes and cancer: a newly described pathway of immune suppressionClin Cancer Res20111795964https://doi.org/10.1158/1078-0432.ccr-10-1489[PubMed][Google Scholar]
  • 365. LeeYEl AndaloussiSWoodMJExosomes and microvesicles: extracellular vesicles for genetic information transfer and gene therapyHum Mol Genet201221R12534https://doi.org/10.1093/hmg/dds317[PubMed][Google Scholar]
  • 366. Al-NedawiKMeehanBMicallefJLhotakVMayLGuhaARakJIntercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cellsNat Cell Biol20081061924https://doi.org/10.1038/ncb1725[PubMed][Google Scholar]
  • 367. SkogJWurdingerTvan RijnSMeijerDHGaincheLSena-EstevesMCurryWTJrCarterBSKrichevskyAMBreakefieldXOGlioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkersNat Cell Biol20081014706https://doi.org/10.1038/ncb1800[PubMed][Google Scholar]
  • 368. DingGZhouLQianYFuMChenJChenJXiangJWuZJiangGCaoLPancreatic cancer-derived exosomes transfer miRNAs to dendritic cells and inhibit RFXAP expression via miR-212-3pOncotarget201562987788https://doi.org/10.18632/oncotarget.4924[PubMed][Google Scholar]
  • 369. SynNWangLSethiGThieryJPGohBCExosome-Mediated Metastasis: From Epithelial-Mesenchymal Transition to Escape from ImmunosurveillanceTrends Pharmacol Sci20163760617https://doi.org/10.1016/j.tips.2016.04.006[PubMed][Google Scholar]
  • 370. PsailaBLydenDThe metastatic niche: adapting the foreign soilNat Rev Cancer2009928593https://doi.org/10.1038/nrc2621[PubMed][Google Scholar]
  • 371. Ordonez-MoranPHuelskenJComplex metastatic niches: already a target for therapy?Curr Opin Cell Biol2014312938https://doi.org/10.1016/j.ceb.2014.06.012[PubMed][Google Scholar]
  • 372. QuailDFJoyceJAMicroenvironmental regulation of tumor progression and metastasisNat Med201319142337https://doi.org/10.1038/nm.3394[PubMed][Google Scholar]
  • 373. HoshinoACosta-SilvaBShenTLRodriguesGHashimotoATesic MarkMMolinaHKohsakaSDi GiannataleACederSSinghSWilliamsCSoplopNTumour exosome integrins determine organotropic metastasisNature201552732935https://doi.org/10.1038/nature15756[PubMed][Google Scholar]
  • 374. ColomboMRaposoGTheryCBiogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesiclesAnnu Rev Cell Dev Biol20143025589https://doi.org/10.1146/annurev-cellbio-101512-122326[PubMed][Google Scholar]
  • 375. RaposoGStoorvogelWExtracellular vesicles: exosomes, microvesicles, and friendsJ Cell Biol201320037383https://doi.org/10.1083/jcb.201211138[PubMed][Google Scholar]
  • 376. YuIMHughsonFMTethering factors as organizers of intracellular vesicular trafficAnnu Rev Cell Dev Biol20102613756https://doi.org/10.1146/annurev.cellbio.042308.113327[PubMed][Google Scholar]
  • 377. KingHWMichaelMZGleadleJMHypoxic enhancement of exosome release by breast cancer cellsBMC Cancer201212421https://doi.org/10.1186/1471-2407-12-421[PubMed][Google Scholar]
  • 378. OstrowskiMCarmoNBKrumeichSFangetIRaposoGSavinaAMoitaCFSchauerKHumeANFreitasRPGoudBBenarochPHacohenNRab27a and Rab27b control different steps of the exosome secretion pathwayNat Cell Biol2010121930https://doi.org/10.1038/ncb2000[PubMed][Google Scholar]
  • 379. BobrieAKrumeichSReyalFRecchiCMoitaLFSeabraMCOstrowskiMTheryCRab27a supports exosome-dependent and -independent mechanisms that modify the tumor microenvironment and can promote tumor progressionCancer Res201272492030https://doi.org/10.1158/0008-5472.can-12-0925[PubMed][Google Scholar]
  • 380. YuXHarrisSLLevineAJThe regulation of exosome secretion: a novel function of the p53 proteinCancer Res2006664795801https://doi.org/10.1158/0008-5472.can-05-4579[PubMed][Google Scholar]
  • 381. LespagnolADuflautDBeekmanCBlancLFiucciGMarineJCVidalMAmsonRTelermanAExosome secretion, including the DNA damage-induced p53-dependent secretory pathway, is severely compromised in TSAP6//Steap3-null miceCell Death Differ200815172333http://www.nature.com/cdd/journal/v15/n11/suppinfo/cdd2008104s1.html[PubMed][Google Scholar]
  • 382. PeinadoHAleckovicMLavotshkinSMateiICosta-SilvaBMoreno-BuenoGHergueta-RedondoMWilliamsCGarcia-SantosGGhajarCNitadori-HoshinoAHoffmanCBadalKMelanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through METNat Med20121888391https://doi.org/10.1038/nm.2753[PubMed][Google Scholar]
  • 383. BalajLLessardRDaiLChoYJPomeroySLBreakefieldXOSkogJTumour microvesicles contain retrotransposon elements and amplified oncogene sequencesNat Commun20112180https://doi.org/10.1038/ncomms1180[PubMed][Google Scholar]
  • 384. RatajczakJMiekusKKuciaMZhangJRecaRDvorakPRatajczakMZEmbryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: evidence for horizontal transfer of mRNA and protein deliveryLeukemia20062084756https://doi.org/10.1038/sj.leu.2404132[PubMed][Google Scholar]
  • 385. Costa-SilvaBAielloNMOceanAJSinghSZhangHThakurBKBeckerAHoshinoAMarkMTMolinaHPancreatic cancer exosomes initiate pre-metastatic niche formation in the liverNat Cell Biol201517https://doi.org/10.1038/ncb3169[Google Scholar]
  • 386. LiPKaslanMLeeSHYaoJGaoZProgress in Exosome Isolation TechniquesTheranostics20177789804https://doi.org/10.7150/thno.18133[PubMed][Google Scholar]
  • 387. MeloSALueckeLBKahlertCFernandezAFGammonSTKayeJLeBleuVSMittendorfEAWeitzJRahbariNGlypican-1 identifies cancer exosomes and detects early pancreatic cancerNature2015523https://doi.org/10.1038/nature14581[Google Scholar]
  • 388. LötvallJHillAFHochbergFBuzásEIDi VizioDGardinerCGhoYSKurochkinIVMathivananSQuesenberryPSahooSTaharaHWaubenMHMinimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular VesiclesJournal of Extracellular Vesicles2014310.3402/jev.v3.26913. https://doi.org/10.3402/jev.v3.26913[Google Scholar]
  • 389. QueRDingGChenJCaoLAnalysis of serum exosomal microRNAs and clinicopathologic features of patients with pancreatic adenocarcinomaWorld J Surg Oncol201311219https://doi.org/10.1186/1477-7819-11-219[PubMed][Google Scholar]
  • 390. KahlertCMeloSAProtopopovATangJSethSKochMZhangJWeitzJChinLFutrealAKalluriRIdentification of Double-stranded Genomic DNA Spanning All Chromosomes with Mutated KRAS and p53 DNA in the Serum Exosomes of Patients with Pancreatic CancerJ Biol Chem2014289386975https://doi.org/10.1074/jbc.C113.532267[PubMed][Google Scholar]
  • 391. MadhavanBYueSGalliURanaSGrossWMullerMGieseNAKalthoffHBeckerTBuchlerMWZollerMCombined evaluation of a panel of protein and miRNA serum-exosome biomarkers for pancreatic cancer diagnosis increases sensitivity and specificityInt J Cancer2015136261627https://doi.org/10.1002/ijc.29324[PubMed][Google Scholar]
  • 392. KanwarSSDunlayCJSimeoneDMNagrathSMicrofluidic device (ExoChip) for on-chip isolation, quantification and characterization of circulating exosomesLab Chip2014141891900https://doi.org/10.1039/c4lc00136b[PubMed][Google Scholar]
  • 393. AllensonKCastilloJSan LucasFASceloGKimDUBernardVDavisGKumarTKatzMOvermanMJForetovaLFabianovaEHolcatovaIHigh prevalence of mutant KRAS in circulating exosome-derived DNA from early-stage pancreatic cancer patientsAnn Oncol2017287417https://doi.org/10.1093/annonc/mdx004[PubMed][Google Scholar]
  • 394. KhanSJutzyJMSAspeJRWallNRAbstract B97: Exosomal survivin, a potential tool for early detection of pancreatic cancer health disparity?Cancer Research201272B97Bhttps://doi.org/10.1158/1538-7445.panca2012-b97[Google Scholar]
  • 395. ShinSJSmithJARezniczekGAPanSChenRBrentnallTAWicheGKellyKAUnexpected gain of function for the scaffolding protein plectin due to mislocalization in pancreatic cancerProc Natl Acad Sci U S A2013110194149https://doi.org/10.1073/pnas.1309720110[PubMed][Google Scholar]
  • 396. PangWSuJWangYFengHDaiXYuanYChenXYaoWPancreatic cancer-secreted miR-155 implicates in the conversion from normal fibroblasts to cancer-associated fibroblastsCancer Sci201510613629https://doi.org/10.1111/cas.12747[PubMed][Google Scholar]
  • 397. TaucherVManggeHHaybaeckJNon-coding RNAs in pancreatic cancer: challenges and opportunities for clinical applicationCell Oncol (Dordr)201639295318https://doi.org/10.1007/s13402-016-0275-7[PubMed][Google Scholar]
  • 398. ZöllerMPancreatic cancer diagnosis by free and exosomal miRNAWorld Journal of Gastrointestinal Pathophysiology201347490https://doi.org/10.4291/wjgp.v4.i4.74[PubMed][Google Scholar]
  • 399. YuCWangMLiZXiaoJPengFGuoXDengYJiangJSunCMicroRNA-138-5p regulates pancreatic cancer cell growth through targeting FOXC1Cell Oncol (Dordr)20153817381https://doi.org/10.1007/s13402-014-0200-x[PubMed][Google Scholar]
  • 400. TianSGuoXYuCSunCJiangJmiR-138-5p suppresses autophagy in pancreatic cancer by targeting SIRT1Oncotarget201781107182https://doi.org/10.18632/oncotarget.14360[PubMed][Google Scholar]
  • 401. RanaSMalinowskaKZollerMExosomal tumor microRNA modulates premetastatic organ cellsNeoplasia20131528195[PubMed][Google Scholar]
  • 402. AliSDubayboHBrandRESarkarFHDifferential Expression of MicroRNAs in Tissues and Plasma Co-exists as a Biomarker for Pancreatic CancerJ Cancer Sci Ther2015733646https://doi.org/10.4172/1948-5956.1000372[PubMed][Google Scholar]
  • 403. MikamoriMYamadaDEguchiHHasegawaSKishimotoTTomimaruYAsaokaTNodaTWadaHKawamotoKGotohKTakedaYTanemuraMMicroRNA-155 Controls Exosome Synthesis and Promotes Gemcitabine Resistance in Pancreatic Ductal AdenocarcinomaScientific Reports2017742339https://doi.org/10.1038/srep42339[PubMed][Google Scholar]
  • 404. OhuchidaKMizumotoKKayashimaTFujitaHMoriyamaTOhtsukaTUedaJNagaiEHashizumeMTanakaMMicroRNA Expression as a Predictive Marker for Gemcitabine Response after Surgical Resection of Pancreatic CancerAnnals of Surgical Oncology20111823817https://doi.org/10.1245/s10434-011-1602-x[PubMed][Google Scholar]
  • 405. van den BoornJGDasslerJCochCSchleeMHartmannGExosomes as nucleic acid nanocarriersAdv Drug Deliv Rev2013653315https://doi.org/10.1016/j.addr.2012.06.011[PubMed][Google Scholar]
  • 406. BellaviaDRaimondoSCalabreseGForteSCristaldiMPatinellaAMemeoLMannoMRaccostaSDianaPCirrincioneGGiavaresiGMonteleoneFInterleukin 3- receptor targeted exosomes inhibit in vitro and in vivo Chronic Myelogenous Leukemia cell growthTheranostics20177133345https://doi.org/10.7150/thno.17092[PubMed][Google Scholar]
  • 407. KharazihaPCederSLiQPanaretakisTTumor cell-derived exosomes: a message in a bottleBiochim Biophys Acta2012182610311https://doi.org/10.1016/j.bbcan.2012.03.006[PubMed][Google Scholar]
  • 408. NakaseIKobayashiNBTakatani-NakaseTYoshidaTActive macropinocytosis induction by stimulation of epidermal growth factor receptor and oncogenic Ras expression potentiates cellular uptake efficacy of exosomesSci Rep2015510300https://doi.org/10.1038/srep10300[PubMed][Google Scholar]
  • 409. CommissoCDavidsonSMSoydaner-AzelogluRGParkerSJKamphorstJJHackettSGrabockaENofalMDrebinJAThompsonCBRabinowitzJDVander HeidenMGMacropinocytosis of protein is an amino acid supply route in Ras-transformed cellsNature20134976337https://doi.org/10.1038/nature12138[PubMed][Google Scholar]
  • 410. KamerkarSLeBleuVSSugimotoHYangSRuivoCFMeloSALeeJJKalluriRExosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancerNature2017546498503https://doi.org/10.1038/nature22341http://www.nature.com/nature/journal/vaop/ncurrent/abs/nature22341.html#supplementary-information[PubMed][Google Scholar]
  • 411. KowalikAKowalewskaMGóźdźSCurrent approaches for avoiding the limitations of circulating tumor cells detection methods—implications for diagnosis and treatment of patients with solid tumorsTranslational Research20171855884.e15https://doi.org/10.1016/j.trsl.2017.04.002[PubMed][Google Scholar]
  • 412. MeachamCEMorrisonSJTumor heterogeneity and cancer cell plasticityNature201350132837https://doi.org/10.1038/nature12624[PubMed][Google Scholar]
  • 413. CorrentiMRaggiCStem-like plasticity and heterogeneity of circulating tumor cells: current status and prospect challenges in liver cancerOncotarget201787094115https://doi.org/10.18632/oncotarget.12569[PubMed][Google Scholar]
  • 414. PixbergCFSchulzWAStoeckleinNHNevesRPCharacterization of DNA Methylation in Circulating Tumor CellsGenes (Basel)20156105375https://doi.org/10.3390/genes6041053[PubMed][Google Scholar]
  • 415. PixbergCFRabaKMullerFBehrensBHonischENiederacherDNeubauerHFehmTGoeringWSchulzWAFlohrPBoysenGLambrosMAnalysis of DNA methylation in single circulating tumor cellsOncogene20173632233231https://doi.org/10.1038/onc.2016.480[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.