Review Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Dec 14, 2022; 28(46): 6478-6496
Published online Dec 14, 2022. doi: 10.3748/wjg.v28.i46.6478
Liquid biopsy leads to a paradigm shift in the treatment of pancreatic cancer
Fumiaki Watanabe, Koichi Suzuki, Hiroshi Noda, Toshiki Rikiyama, Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
ORCID number: Fumiaki Watanabe (0000-0003-2944-0985); Koichi Suzuki (0000-0002-3548-0628); Hiroshi Noda (0000-0002-3514-1706); Toshiki Rikiyama (0000-0002-9417-7020).
Author contributions: All authors contributed to the design of the study. Watanabe F and Suzuki K drafted the manuscript and analyzed data; All other authors contributed to the manuscript review; The final version of the manuscript was read and approved by all authors.
Supported by Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology, No. JP 16K10514; and the JKA Foundation through its promotion funds from the Keirin Race, No. 27-1-068 (2).
Conflict-of-interest statement: Authors declare no conflict of interests for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Fumiaki Watanabe, MD, PhD, Assistant Professor, Doctor, Department of Surgery, Saitama Medical Center, Jichi Medical University, 1-847, Amanuma-cho, Minuma-ku, Saitama 330-8503, Japan. fwatanabe@jichi.ac.jp
Received: September 20, 2022
Peer-review started: September 20, 2022
First decision: October 18, 2022
Revised: October 25, 2022
Accepted: November 21, 2022
Article in press: November 21, 2022
Published online: December 14, 2022

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is one of the most cancers. Its 5-year survival rate is very low. The recent induction of neoadjuvant chemotherapy and improvements in chemotherapy for patients with pancreatic cancer have resulted in improved survival outcomes. However, the prognosis of pancreatic cancer is still poor. To dramatically improve the prognosis, we need to develop more tools for early diagnosis, treatment selection, disease monitoring, and response rate evaluation. Recently, liquid biopsy (circulating free DNA, circulating tumor DNA, circulating tumor cells, exosomes, and microRNAs) has caught the attention of many researchers as a new biomarker that is minimally invasive, confers low-risk, and displays an overall state of the tumor. Thus, liquid biopsy does not employ the traditional difficulties of obtaining tumor samples from patients with advanced PDAC to investigate their molecular biological status. In addition, it allows for long-term monitoring of the molecular profile of tumor progression. These could help in identifying tumor-specific alterations that use the target structure for tailor-made therapy. Through this review, we highlighted the latest discoveries and advances in liquid biopsy technology in pancreatic cancer research and showed how it can be applied in clinical practice.

Key Words: Pancreatic cancer, Cell-free DNA, Circulating tumor DNA, Circulating tumor cells, Exosomes, MicroRNAs

Core Tip: We focused on liquid biopsy technology for pancreatic ductal adenocarcinoma (PDAC), including circulating free DNA, circulating tumor DNA, circulating tumor cells, exosomes, and microRNAs. We equally described the characteristics of these technologies and reviewed the clinical significance according to the purpose of these biomarkers: early diagnosis, prognosis, prediction of recurrence, and therapeutic response. Although liquid biopsy still has many limitations to its widespread utilization in clinical practice, liquid biopsy has the potential to be applied from diagnosis to treatment. It is expected to improve the prognosis of PDAC radically.



INTRODUCTION

Pancreatic ductal adenocarcinoma (PDAC) is the seventh most common cause of cancer-related death worldwide, and its incidence is increasing[1-3]. In the United States and Japan, it ranks as the fourth leading cause of cancer-related mortality[4,5]. Its 5-year survival rate remains as low as 6% in the United States[6]. Without novel diagnostic methods and/or treatments, it is expected to become the second leading cause of cancer-related deaths by 2030[7]. Due to its early metastatic nature, up to 20% of patients with PDAC are eligible for initial resection[6]. The poor prognosis results from the low resectability rate at diagnosis, with surgery being the only potentially curative treatment. However, even with radical resection, most patients relapse within a year. Moreover, due to the high resistance rate to chemotherapy, radiotherapy, and immunotherapy[8], non-operative treatment has a poorer prognosis with a median survival of 5-9 mo[9].

A variety of genetic and molecular alterations have been identified in PDAC, including mutations in KRAS, p16, p53, BRCA2, Smad4, etc[10]. However, translating this scientific knowledge into clinical treatment regimens has not yet been achieved. Tumor marker-adjusted treatments for PDAC have been discussed by several authors[11-16]. Carbohydrate antigen 19-9 (CA19-9) is considered the best tumor marker for patients with PDAC. CA19-9 values correlate with tumor size, stage, and burden[17,18]. Therefore, CA19-9 is commonly used to diagnose PDAC, assess resectability, monitor progression, and determine prognosis[19]. Preoperative and postoperative CA19-9 levels predict prognosis[20-24]. Although CA19-9 is a helpful prognostic factor of PDAC, its usefulness remains controversial[19,25-27]. Better tumor markers that correlate with tumor size, predict recurrence after surgery, reflect tumor progression and metastasis, and indicate response to chemotherapy are urgently needed to improve the prognosis of PDAC.

This ideal tumor biomarker should be disease-specific and sensitive. It should also have a high positive predictive value and allows for the detection of disease at very early stages. Furthermore, it should be easy to collect, and the test should be simple and economically feasible. To replace CA19-9 as an alternative biomarker, many researchers have reported the usefulness of liquid biopsy in various cancers. Liquid biopsy is used to diagnose cancer through the detection of circulating tumor cells (CTCs), cell-free and circulating tumor DNA (cfDNA and ctDNA), and microvesicles such as exosomes containing nucleic acids and proteins released from primary tumors and metastases into body fluids. In contrast to surgical or needle biopsy, liquid biopsy is a non-invasive diagnostic method. As a result, it can be detected in real-time and can provide valid information[28-33].

Through this review, we aimed at exploring cfDNA, ctDNA, CTCs, exosomes, and microRNAs (miRNAs) in liquid biopsy technology for early detection, prognostic evaluation, prediction of response to chemotherapy, development of acquired resistance, and early detection of disease relapse, and to evaluate their clinical utility.

CFDNA

cfDNA in plasma possibly originates from necrosis, apoptosis, and/or macrophage digestion of tumoral and healthy cells. Previous studies demonstrated that most of the plasma cfDNA molecules originate from the hematopoietic system in healthy individuals[34,35]. However, in certain physiological or pathological conditions, such as pregnancy, organ transplantation, and cancers, the related/affected tissues could release additional DNA into peripheral circulation[36-38]. In 1948, Mandel et al[39] first discovered circulating cfDNA. However, it was not until 1977 that the utility of cfDNA was appreciated when Leon et al[36] discovered that significantly elevated levels were detected in patients with cancer compared to healthy controls. In 1989, Stroun et al[40] found that some of the cfDNA in the plasma of cancer patients was derived from cancer cells (called ctDNA). Subsequent studies revealed that cancer cells release ctDNA fragments into blood and other biological fluids such as urine, saliva, and cerebrospinal fluid. ctDNA is highly fragmented, with a shorter fragment length found in cancer patients (134-145 bp) than that of healthy individuals (165-167 bp)[41]. ctDNA is derived from various tumor sites and can provide much more comprehensive genomic and epigenomic information than single-site biopsies. Thus, ctDNA overcomes the issue of tumor heterogeneity, a significant limitation of conventional tissue biopsy. Furthermore, its non-invasive nature allows for continuous real-time monitoring of the molecular status of cancer.

CTCS

CTCs are defined as cells derived from primary, recurrent, or metastatic tumors. CTCs were reported in 1869, and their significance in peripheral blood has been extensively studied in various malignancies[42-46]. It potentially correlated with tumor metastasis and recurrence in breast cancer[42,43], prostate cancer[44], lung cancer[45], and colorectal cancer[46]. Multiple techniques have been developed to explore CTCs, tumor-specific epitopes that are not present in normal blood cells, changes in physical properties such as size, density, and electromechanical properties, or high-throughput imaging to uncollected blood cell preparations[47]. CTCs can travel in the interstitium and bloodstream as single cells as well as clusters[48,49]. Several studies have depicted that CTC clusters correlate with high metastatic potential and poor outcomes. CTC clusters were more significantly associated with distant metastasis than single CTCs[50,51]. Its predictive value has been explored in recent studies in patients with lung, breast, and colon cancer[48,52,53].

EXOSOMES

In 1967, Peter Wolf first discovered that platelets release numerous vesicles[54]. At the time, these vesicles were regarded as cell fragments with no associated biological function. In 1983, exosomes were first observed in sheep reticulocytes, and Johnstone named them "exosomes" in 1987[55]. They are one of the extracellular vesicles (EVs) and have attracted attention in tumor biology recently. Exosomes are 40-150 nm in diameter containing transmembrane proteins, heat shock proteins, nucleic acids (DNA, mRNA, miRNA, long and short non-coding RNA), and enzymes (GAPDH, ATPase, pgk1, RAB)[56-58]. The molecular content of exosomes reflects the nature and state of the cell from which they originate, and their content can alter the function of the recipient cell[59]. In 1996, B lymphocyte-derived exosomes were found to exhibit antigen-presenting properties and induce T-cell responses[60]. Similarly, antigen-presenting exosomes derived from dendritic cells retarded the progression of cancer[61]. These lipid-bilayer vesicles apparently regulate tumor drug resistance, metastasis, and suppression of immune responses. From tumor growth to cellular metastasis, a complex exosome communication network between tumor and non-tumor cells directs all stages[62]. Tumor cells develop exosome-based mechanisms that promote a favorable microenvironment to support tumor growth by facilitating cell metastasis, evading apoptosis, establishing a premetastatic niche, and transporting information from cell to cell[62]. Tumor-derived exosomes can efficiently be captured by various separation methods and gives substantial information about the tumor. In addition, since exosomes are ubiquitously present in body fluids, diffuse through them, and fuse with cell membranes to exert their effects, they are potential tumor therapeutic drug carriers.

MIRNAS

miRNAs are non-coding RNAs that are involved in regulating gene expression. Most miRNAs are transcribed from DNA sequences to become primary miRNAs, then processed into precursor miRNAs and finally mature miRNAs. In most cases, miRNAs interact with 3′ untranslated region (3′ UTR) of the target mRNA to induce mRNA degradation and translational repression. However, interactions with other regions, such as the 5′ UTR, coding sequences, and gene promoters, have also been reported. Under certain conditions, they activate translation or regulate transcription. The interaction of miRNAs with their target genes is dynamic. It depends on many factors, including the intracellular location of the miRNA, the amount of miRNA and target mRNA present, and the affinity of the miRNA-mRNA interaction. MiRNAs are secreted into the extracellular fluid and transported to target cells via vesicles such as exosomes or binding to proteins such as Argonaute. Extracellular miRNAs mediate cell-to-cell communication as chemical messengers. The first miRNAs were reported in the nematode, C. elegans by the Ambros group at Harvard University in 1993[63]. Ambros et al[63] screened for mutants that affect the timing of cell fate switching during nematode development. They identified two genes: lin-4 and let-7. Surprisingly, these genes did not code for proteins but for small RNAs, later called miRNAs. So far, over 38000 miRNAs have been identified and catalogued in the public database, miRBase (www.mirbase.org). miRNAs are involved in a wide range of processes, including metabolism, cell proliferation, apoptosis, and developmental timing[64]. Overexpressed miRNAs act as regulators of oncogenes (through downregulation of tumor suppressor genes) and/or cellular processes such as cell differentiation and apoptosis. Thus, miRNAs are associated with the development of various types of cancer, including colorectal, breast, ovarian, and endometrial cancer[65-68], but their exact pathways are not fully understood. MiRNAs are involved in cell growth and differentiation regulators and have been proposed to be good candidates for diagnosis and treatment of cancers[69].

CLINICAL SIGNIFICANCE OF LIQUID BIOPSY IN PANCREATIC CANCER
Early diagnosis

KRAS mutations in tumoral tissues have been detected in 90% of PDAC[9,70,71] and the heterogeneity of KRAS mutations between primary tumor and metastasis in patients with PDAC[72,73]. Due to high penetration of KRAS mutations in PDAC patients, identification of KRAS mutations in cfDNA could be a suitable cancer biomarker. CfDNA, however, is immediately removed from the circulatory system by nuclease action and urinary excretion. In addition, uptake by the liver and spleen and degradation by macrophages may also affect its removal from the circulation[74,75]. This short half-life, a few hours at most[74], makes it complicated to detect cfDNA and ctDNA in early-stage cancer.

CTCs was not highly observed in early-stage PDAC, with a sensitivity of < 50%[76,77]. The detection rate of CTCs is 0.0%, 60.7%, 78.6%, and 96.3% of American Joint Commission on Cancer stages I, II, III, and IV patients, respectively, and increases dependence on cancer progression[76]. On the other hand, CTCs have also been detected in patients with early-stage PDAC[78-80], with relatively high sensitivity of over 70%. Kulemann et al[81] reported that no difference in the detection rate of CTCs between early-stage and advanced PDAC, which may suggest that CTCs are disseminated from the primary tumor in the early stages of the disease and may be used to diagnose PDAC in the initial stages.

Exosomes have shown promise for early detection of PDAC and proved to be a useful tool clinically[33,82]. Melo et al[33] found glypican-1 (the cell surface proteoglycan glypican-1) on tumor-derived exosomes as a diagnostic biomarker in PDAC, which enabled to distinguish healthy persons from patients with benign diseases and patients with early- and late-stage pancreatic cancer with 100% of the sensitivity and specificity. Another study, however, failed to show significant differences in GPC1 between healthy, PDAC and chronic pancreatitis samples while a combination of detection with high levels of exosomal miR-10b, miR-21, miR-30c, and miR-181a and low levels of miR-let7a succeeded to distinguish PDAC from healthy and chronic pancreatitis samples[83]. The EV is also a potent biomarker for early detection. Liang et al[84] identified ephrin type-A receptor 2 (EphA2) to distinguish PDAC patients from pancreatitis patients and healthy control. In addition, it discriminated against PDAC patients with early disease (stage I/II), who were potentially benefit from curative surgical resection, from normal healthy control (NC) and pancreatitis cases. The blood levels of EphA2-EV before treatments distinguished stage I/II PDAC patients from NC and pancreatitis accurately.

Differences in expression of miRNA are considered precursors to prominent diagnostic biomarkers of PDAC[85-89]. High expressions of serum exosomal miR-17-5p and miR-21 are likely observed in PDAC patients in connection with metastasis and the advanced stage of PDAC[85]. Studies of miR-10b[86,87] also showed increased levels in exosomes isolated from the plasma of PDAC patients compared to those with chronic pancreatitis or normal controls. Peng et al[90] conducted the meta-analysis including 46 studies and found that the sensitivity, specificity, and AUC of circulating miRNAs for discriminating early-stage PDAC patients (0-IIa) from non-PDAC controls were 0.78 (0.76-0.81), 0.78 (0.75-0.80) and 0.85 (0.82-0.88), respectively.

cfDNA, ctDNA, and CTCs are highly detectable in patients with advanced PDAC, but less noticeable in early-stage PDAC while exosomes are secreted into cells from early stages and miRNAs are abundant and relatively easy to detect. These biomarkers, however, are not yet appropriate for early diagnosis in clinical practice and liquid biopsy (especially cfDNA and ctDNA) needs to be improved for the detection capability as a screening tool for PDAC (Table 1).

Table 1 Liquid biopsy in the early diagnosis of pancreatic cancer.
Ref.
Journal
No. of patients
Biomarker
Method
Main findings
Ankeny et al[76], 2016Br J Cancer72CTCMicrofluidic NanoVelcro CTC chipDetection rate of PDAC: 54/72 (sensitivity = 75.0%, specificity = 96.4%)
Rhim et al[77], 2014Gastroenterology51CTCMicrofluidic geometrically enhanced differential immunocaptureCTC (≥ 3) in 33% of patients with cystic lesions and no clinical diagnosis of cancer, 73% with PDAC, and 0% of controls
Xu et al[78], 2017Int J Mol Sci40CTCNE-iFISHThe positive rate of diagnosis of PDAC is nearly 97% in combination with CA19-9
Poruk et al[79], 2017Clin Cancer Res60CTCISET method & immunofluorescence The positive rate is 12% in stageⅠPDAC
Gao et al[80], 2016J Exp Clin Cancer Res25CTCSE-iFISH platformSensitivity of 88 % and specificity of 90 % in PDAC
Kulemann et al[81], 2015Pancreas11CTCScreenCell; Cyto kitNo difference in the rate of CTC detection between early-stage and advanced-stage diseases (P = 0.71)
Melo et al[33], 2015Nature56ExosomeFACS analysisThe sensitivity and specificity of GPC1+ circulating exosomes in diagnosing PDAC were both 100%
Buscail et al[82], 2019Cancers30Exosome and CTCFACS, Cellsearch and RosetteSepTMCombining quantification of GPC1-positive exosomes and CTC detection identified all the PDAC patients, showed a negative predictive value of 100%, and an overall diagnostic accuracy of 91%
Lai et al[83], 2017Cancer Lett40Exosome and miRNALC-MS & RT-qPCRHigh levels of exosomal miR-10b, miR-21, miR-30c, and miR-181a and low levels of miR-let7a differentiated PDAC from healthy and chronic pancreatitis samples
Liang et al[84], 2017Nat Biomed Eng23EVnPES assayPre-therapy EphA2-EV blood levels accurately distinguished stage I/II pancreatic cancer patients from NC (AUC = 0.96) and pancreatitis patients (AUC = 0.93)
Que et al[85], 2013World J Surg Oncol49miRNART-PCRSerum exosomal miR-17-5p was higher in PDAC patients than in non–PDAC patients and healthy participants
Cote et al[86], 2014Am J Gastroenterol215miRNART-PCRIncreased expression of miRNA-10b, -155, and -106b in plasma appears highly accurate in diagnosing PDAC
Ouyang et al[87], 2015Oncogene42miRNART-PCRPlasma miR-10b levels significantly increased in comparison with normal controls
Slater et al[88], 2014Transl Oncol59miRNAReal-time PCRA combination test of miRNA-196a and miRNA-196b, whose expression is upregulated from the PanIN state, can identify patients with PanIN 2/3
Madhavan et al[89], 2015Int J Cancer220Exosome and miRNAmiRNeasyMinikit, RT-PCR, qRT-PCR and flow cytometryThe selected miR-1246, miR-4644, miR-3976 and miR-4306 were significantly upregulated in 83% of PDAC serum-exosomes, but rarely in control groups
Prognosis assessment

Numerous articles have been published regarding the prognosis of PDAC using liquid biopsy. It is challenging to detect cfDNA in early-stage PDAC due to small amount of cfDNA at the instance. On the other hand, cfDNA is detected with high probability in plasma from patients with advanced PDAC and high levels of cfDNA are significantly associated with poor prognosis[91-93]. Higher levels of plasma DNA (> 62 ng/mL) is significantly associated with poor outcome in overall survival (OS), presence of vascular encasement and metastasis[91]. Fragment size of cfDNA and cfDNA are investigated in patients with advanced PDAC, where a pretreatment cfDNA fragment size of 167 bp or less and a high pretreatment cfDNA level are associated with shorter progression-free survival (PFS) and OS[92]. Regarding ctDNA, KRAS mutations in plasma from PDAC patients were detected in 1999 in connection with poor survival[94]. Then, several studies reported the feasibility of detecting circulating mutant KRAS genes in the blood of PDAC patients, as well as the prognostic relevance of KRAS genes[95-103]. Hadano et al[96] assessed the prognosis of patients who underwent curative pancreatoduodenectomy for PDAC according to the presence of KRAS-mutated ctDNA before surgery. The median OS of patients with PDAC with ctDNA was significantly worse than that of those without ctDNA (13.6 and 27.6 mo, respectively). ctDNA before surgery is not associated with prognosis or recurrence[97,98]. Bernard et al[99] validated the significance of change in ctDNA and exosome DNA (exoDNA) during neoadjuvant chemotherapy in resectable PDAC patients and found that increased exoDNA levels after neoadjuvant chemotherapy were significantly associated with disease progression, whereas ctDNA showed no correlation with outcome. They also elucidated the significance of the presence of ctDNA and exoDNA before chemotherapy in unresectable PDAC patients demonstrating that detection of ctDNA and exoDNA mutant allele frequency (MAFs) ≥ 5% at baseline status were a significantly poor prognosis. Kinugasa et al[100] reported that KRAS mutations were observed in 62.5% of serum samples of 75 patients with PDAC at all stages using droplet digital polymerase chain reaction (PCR) and were correlated with worse OS. KRAS mutations in ctDNA were an independent negative predictor of survival in unresectable pretreatment PDAC patients[101-103].

CTCs serve as prognostic markers in several studies. A recent 9-cohort meta-analysis of separate studies using CellSearch and reverse transcription PCR detection methods involving 623 PDAC patients found an association between detection of CTCs and poor prognosis. Among the 623 patients, 268 patients (43%) with CTCs showed poor PFS and OS compared to the those without CTCs[104]. The different methods of enrichment and detection of CTCs demonstrate that the abundance of CTCs[76,105-108], circulating tumor microemboli (CTMs)[109], could be predictive of worse survival. Results have varied depending on the isolation techniques, detection methods, and the population of patients. Bidard et al[110] investigated the CTC detection rate using CellSearch® in a subgroup of 79 patients with locally advanced PDAC enrolled in the LAP 07 trial; 11% with CTCs had worse OS. Using the CellSearch enrichment method, Kurihara et al[111] investigated the significance of CTCs as a biomarker of clinical outcomes in 26 patients. Eleven of 26 patients (42%) showed CTCs. PDAC patients without CTCs exhibited significant longer median survival times of 375.8 d than those with CTCs (110.5 d, P < 0.001). de Albuquerque et al[112] also reported a worse median PFS in patients with CTC (47% of patients). Detection of CTCs using immunomagnetic epithelial cell adhesion molecule and mucin1 demonstrated longer PFS of 138.0 d in patients without CTCs than in those with CTC (66.0 d). Intriguingly, CTC enumeration is not correlated with clinicopathological features of the disease, including metastasis status and tumor stages. The detection of CTCs alone makes no difference in the prognosis of PDAC patients[113].

The exosomal miRNA-mediated cell-to-cell signaling in the tumor microenvironment plays a significant role in the progression of cancer[114,115]. The proliferation and invasive properties of surrounding cancer cells were shown to be modulated by PDAC exosomal miR-222[116], which was related to poor outcome in PDAC patients[116]. PDAC cells releases exosomes enriched with miR-301a under hypoxic conditions, where circulating exosomal miR-301a-3p levels were positively associated with depth of invasion, lymph node metastasis, late TNM stage, and poor prognosis of PDAC patients[117]. Exosomal miRNAs have been studied to assess PDAC growth, migration, and invasion; further longitudinal studies are essential to identify exosomal miRNAs as prognostic biomarkers for PDAC. Exosomal proteins also play a significant role in PDAC diagnosis. The levels of GPC1 are associated with tumor size and disease burden of PDAC[118]. Macrophage migration inhibitory factor (MIF) is highly expressed in PDAC-derived exosomes and its blockade prevented liver pre-metastatic niche formation and metastasis[119]. MIF was markedly higher in exosomes from patients of stage I PDAC, who later developed liver metastasis, in comparison with advanced PDAC patients, suggesting that increased levels of exosomal MIF could be a biomarker for developing liver metastasis in PDAC patients (Table 2).

Table 2 Liquid biopsy in the predicting prognosis of pancreatic cancer.
Ref.
Journal
year
No. of patients
Biomarker
Method
Main findings
Singh et al[91], 2015Cancer Invest2015cfDNAHigher level of plasma DNA (> 62 ng/mL) was found to associate significantly with lower overall survival time (P = 0.002), presence of vascular encasement (P = 0.030) and metastasis (P = 0.001)
Lapin et al[92], 2018J Transl Med201861cfDNA2100 BioanalyzerPre-treatment cfDNA levels could independently predict prognosis for both PFS (HR = 3.049, P = 0.005) and OS (HR = 2.236, P = 0.028)
Wang et al[93], 2021Pancreas202197cfDNAPCRThe 1- and 5-year survivals for those with high cfDNA were poorer; 70.2% and 21.2%, respectively, as compared with 93.4% and 23.7% for those with low cfDNA level
Castells et al[94], 1999J Clin Oncol199947ctDNAPCR-RFLP and SSCPPlasma KRAS mutations were identified as the only independent prognostic factor (odds ratio, 1.51; 95%CI: 1.02 to 2.23)
Ako et al[95], 2017Pancreatology201740ctDNAddPCRKRAS mutation at G12V in the plasma or serum conferred a significantly poorer prognosis than without the mutation (P < 0.01)
Hadano et al[96], 2016Br J Cancer2016105ctDNAddPCRPatients who were preoperative ctDNA+ had a significantly poorer prognosis with respect to OS (P < 0.0001)
Nakano et al[97], 2018Br J Cancer201845ctDNAPNA directed, PCR clampingThere were no significant differences in DFS and OS between patients with and without KRAS mutations from preoperative serum
Watanabe et al[98], 2019PLoS One201978ctDNAddPCRNo effect of the presence of KRAS-mutated ctDNA before surgery on RFS (median: 16.9 mo vs 32.4 mo) was observed
Bernard et al[99], 2019Gastroenterology201934ctDNA and exosome DNAddPCRIncreased exosome DNA levels after neoadjuvant therapy were significantly associated with disease progression (P = 0.003)
Kinugasa et al[100], 2015Cancer201575ctDNAddPCRKRAS mutations in plasma correlated with poor OS (P = 0.002)
Tjensvoll et al[101], 2016Mol Oncol201614ctDNAPNA clamp PCRKaplan-Meier survival analyses indicated that patients with a positive ctDNA before or after initiation of chemotherapy had shorter PFS and OS
Chen et al[102], 2010Eur J Surg Oncol201091ctDNADirect sequencingKRAS codon 12 mutation from plasma DNA was an independent negative prognostic factor (HR, 7.39; 95%CI: 3.69-14.89)
Sausen et al[103], 2015Nat Commun2015101ctDNANext-generation sequencing and digital PCRctDNA was an independent prognostic marker of OS in advanced disease, with OS of 6.5 mo vs 19.0 mo for ctDNA-positive and negative patients, respectively
Khoja et al[105], 2012Br J Cancer201254CTCCellsearch and ISETThe PFS and OS for patients without vs those with CTCswas 140 d vs 94 d (P = 0.13) and 164 d vs 127 d (P = 0.26), respectively
Earl et al[106], 2015BMC Cancer201545CTCCellsearchA Cox regression analysis showed a significant difference in OS for CTC positive vs negative patients with a HR of 3.0 (P = 0.023)
Zhang et al[107], 2015Int J Cancer201561CTCThe EpCAM-independent methodCTCs positive pancreatic cancer patients exhibit a worse (P = 0.0458) survival rate
Okubo et al[108], 2017Eur J Surg Oncol201765CTCCellsearchA multivariate analysis identified the presence or absence of CTCs as an independent prognostic factor (P = 0.049)
Ankeny et al[76], 2016Br J Cancer2016100CTCMicrofluidic NanoVelcro CTC chipA cut-off of 3 CTCs in 4 mL venous blood was able to discriminate between local/regional and metastatic disease (AUROC = 0.885; 95%CI: 0.800-0.969; and P < 0.001)
Chang et al[109], 2016Clin Chem201663CTManti-EpCAM conjugated supported lipid bilayer-coated microfluidic chipsCTM was an independent prognostic factor of OS and PFS (P 0.0001 and P = 0.003, respectively)
Bidard et al[110], 2013Ann Oncol201379CTCCellsearchCTC positivity was associated with poor tumor differentiation (P = 0.04), and with shorter OS in multivariable analysis (P = 0.01)
Kurihara et al[111], 2008J Hepatobiliary Pancreat Surg200847CTCCellsearchMST of the CTC-positive and -negative patients were 110.5 and 375.8 d (P < 0.001)
de Albuquerque et al[112], 2008Oncology201274CTCMedian PFS time was 66.0 d for patients with baseline CTC positivity and 138.0 days for CTC-negative patients (P = 0.01)
Kulemann et al[81], 2015Pancreas201521CTCScreenCellThe presence of CTC did not adversely affect MST: 16 mo in CTC-positive (n = 18) vs 10 mo in CTC-negative (n = 3) patients
Li et al[116], 2018Cell Physiol Biochem201873miRNAArraystar Human miRCURYTM LNA ArrayMultivariate analyses showed that exosomal miR-222 was independent risk factors for PDAC survival (P = 0.046)
Wang et al[117], 2018Cancer Res201850miRNAqRT-PCRExosomal miR-301a-3p overexpression predicted late TNM stage and poor survival in human PDAC (P = 0.0182)
Frampton et al[118], 2018Oncotarget201843GPC1+ circulating exosomesELISAPatients with high crExos GPC1 levels have significantly larger PDACs (> 4 cm; P = 0.012)
Costa-Silva et al[119], 2015Nat Cell Biol201555ExosomeELISAIncreased levels of MIF in exosomes isolated from patients with PDAC with progression of disease post-diagnosis compared with PDAC patients with no evidence of disease five years post-diagnosis (P < 0.01) and with healthy controls (P < 0.01), but not patients with liver metastasis
Recurrence monitoring

A liquid biopsy is a promising tool to detect minimal residual disease in various gastrointestinal malignancies. Predicting cancer recurrence earlier improves prognosis and provides more options for early consideration of anticancer drugs and operations. Longitudinal monitoring of ctDNA demonstrated to show tumor dynamics in colorectal cancer due to its short half-life[74], which could be available for PDAC.

The monitoring of ctDNA in PDAC patients after surgery reflects early recurrence[97,98,103,120-123]. Multivariate analysis revealed that detection of KRAS mutations in postoperative serum was an independent prognostic factor for disease-free survival (DFS), which is associated with recurrence (P = 0.027)[97]. Increase of KRAS mutated ctDNA after surgery, during periods of adjuvant chemotherapy and observation, was a highly predictive dynamic marker of early relapse[120]. We reported that detection of KRAS mutated ctDNA after surgery was associated with OS regardless of recurrence (P = 0.005) while increase of CA19-9 was associated with recurrence (P < 0.001) but not OS (P = 0.692)[98]. Yamaguchi et al[124] reported that detection of ctDNA before surgery was significantly associated with poor DFS. Detection of ctDNA before surgery is likely to be associated with recurrence comparing to the monitoring of ctDNA after surgery[125-128]. KRAS mutated ctDNA before surgery, especially KRAS G12D mutation, serves as a biomarker for early relapsein resectable PDAC patients[125]. Lee et al[126] reported that KRAS mutated ctDNA before and after surgery was associated with shorter recurrence-free survival in resectable PDAC patients and all patients with ctDNA had a recurrence. Recurrence after surgery is likely identified earlier by ctDNA than imaging study of computed tomography[103,121], suggesting that ctDNA may serve as postoperative surveillance.

While preoperative CTCs have been reported to predict tumor recurrence, postoperative CTC monitoring has been rarely evaluated. Park et al[129] reported that 12 of 40 patients with CTC (33.3%) in preoperative blood showed a significantly frequent rate of systemic recurrence (distant metastases and peritoneal dissemination). Multivariable logistic regression analysis showed the detection of CTC was an independent risk factor for early recurrence and systemic recurrence.

The detection of exosome-derived KRAS with MAFs of > 1% is associated with worse DFS after resection in patients with localized PDAC[130]. Exosomal miR-451a in plasma of PDAC patients is linked to recurrence after surgery[131]. miRNA derived from portal vein blood exosomes (miR-4525, miR-451a, and miR-21), as well as CTCs, can be utilized for the evaluation of PDAC recurrence[132] (Table 3).

Table 3 Liquid biopsy in recurrence monitoring of pancreatic cancer.
Ref.
Journal
No. of patients
Biomarker
Method
Main findings
Nakano et al[97], 2018Br J Cancer45ctDNAPNA-directed PCR clampingMultivariate analysis revealed that KRAS mutations in postoperative serum are an independent prognostic factor for DFS (P = 0.027). Furthermore, the change from not detecting mutant KRAS in preoperative to mutant KRAS in postoperative cfDNA was an independent prognostic factor for OS (P = 0.004)
Hussung et al[120], 2021BMC Cancer25ctDNAddPCR, PCRAn increased KRAS mutated ctDNA during adjuvant chemotherapy and follow-up was a highly predictive dynamic marker of early relapse and poor OS
Watanabe et al[98], 2019PLoS One78ctDNAddPCRDetection of mutant KRAS on postoperative ctDNA was associated with OS regardless of recurrence (P = 0.005)
Groot et al[121], 2019Clin Cancer Res59ctDNAddPCRctDNA detected during follow-up predicted clinical recurrence (sensitivity 90%, specificity 88%) with a median lead time of 84 d
Sausen et al[103], 2015Nat Commun20 (surgery group)ctDNANext-generation sequencing and digital PCRPatients with detectable ctDNA after surgical resection (n = 10) were more likely to relapse and die from disease compared with those with undetectable ctDNA (P = 0.0199)
Jiang et al[122], 2020Front Oncol27ctDNANext-generation sequencingPatients with ctDNA-positive status postoperatively had a markedly reduced DFS compared to those with ctDNA-negative status (P = 0.019)
Kim et al[123], 2018Clin Chem106ctDNAddPCRPatients who had increased KRAS MAF values at 6 mo had a shorter OS (P = 0.036) than those who had decreased values
Yamaguchi et al[124], 2021Ann Surg Oncol97ctDNAddPCRThe multivariate analysis showed that the presence of preoperative ctDNA was associated with poorer OS (P = 0.008) and that postoperative ctDNA was not associated with either RFS or OS
Guo et al[125], 2020Br J Cancer113 and 44 (discovery and validation cohorts)ctDNAddPCRSurvival analysis showed that plasma KRAS mutations, especially KRAS G12D mutation, had significant association with OS and RFS of resectable PDAC. Plasma KRAS G12D mutation showed a strong correlation with early distant metastasis
Lee et al[126], 2019Ann Oncol42ctDNAPCR-based-SafeSeqS assaysPreoperative ctDNA detection was associated with inferior RFS (P = 0.002) and OS (P = 0.015). Detectable ctDNA following curative intent resection was associated with inferior RFS (P < 0.0001) and OS (P = 0.003)
Pietrasz et al[127], 2017Clin Cancer Res31ctDNANext-generation sequencingThe presence of ctDNA was associated with a shorter DFS (4.6 mo vs 17.6 mo; P = 0.03) and shorter OS (19.3 mo vs.32.2 mo; P = 0.027)
Okada et al[128], 2020J Gastroenterol66 (surgery group)ctDNADigital PCRPatients with preoperative ctDNA MAF > 0.45% exhibited significantly shorter disease-free survival than those with lower MAF (HR 3.179, 95%CI: 1.025-9.859; P = 0.0452)
Park et al[129], 2021Sci Rep40CTCCD-PRIM kitOn multivariable logistic regression analysis, CTC positivity was an independent risk factor for early recurrence (P = 0.027) and systemic recurrence (P = 0.033)
Allenson et al[130], 2017Ann Oncol142 and 121 (discovery and validation cohort)Exosome and ctDNAElectron microscopy, flow cytometry and particle analysis and ddPCRHigher exosome KRAS MAFs were associated with decreased disease-free survival in patients with localized disease (P = 0.031)
Takahasi et al[131], 2018J Hepatobiliary Pancreat Sci50miRNAqRT-PCRIn cox proportional hazards model analysis, exosomal miR-451a showed significance to OS and DFS (P = 0.001, P = 0.004)
Kawamura et al[132], 2019J Hepatobiliary Pancreat Sci55miRNAqRT-PCRmiR-4525, miR-451a, and miR-21 from portal vein can be utilized for the evaluation of pancreatic cancer recurrence (P = 0.002, 0.001 and 0.002, respectively)

Therapeutic effect monitoring

An accurate real-time understanding of tumor dynamics during chemotherapy helps to select adequate drugs and avoid unnecessary side effects. For this purpose, more sensitive and novel biomarkers are required to overcome disadvantages of conventional biomarkers including CA19-9; however, only few studies have described monitoring using liquid biopsy during chemotherapy in unresectable PDAC, and most of them reported that the prognosis is poor when each marker is detected[78,84,98,99,108,133-137].

The importance of long-term monitoring of ctDNA its predictive ability for the prognosis of PDAC[98,133,134]. In 2018, Kruger et al[134] highlighted the cut-off value of ctDNA for detecting early drug response in metastatic PDAC patients who underwent chemotherapy: an increase in ctDNA at day 14 correlated with disease progression determined by subsequent imaging study with a sensitivity of 83% and specificity of 100%. Yin et al[138] evaluated ctDNA in patients with PDAC with pathologic complete response (pCR) to neoadjuvant chemotherapy and found its associations with the outcome. They reported that ctDNA existed even in patients with PDAC with pCR to neoadjuvant chemotherapy, thereby predicting early recurrence and reduced survival. These data were obtained using somatic mutations in tumor tissues and CTCs in addition to ctDNA.

Change in number of CTCs was reported in response to neoadjuvant chemotherapy in patients with advanced stage PDAC[138-140]. In a study of 57 PDAC patients who underwent surgery, patients who received neoadjuvant chemotherapy had significantly lower number of CTC than those who were chemotherapy-naive at the time of surgery[139]. Another study, however, demonstrated no differences in 16 patients who received neoadjuvant chemotherapy[140]. Wei et al[135] evaluated CTCs in patients with PDAC who underwent surgery and chemotherapy (modified FOLFIRINOX; oxaliplatin, leucovorin, irinotecan, and fluorouracil, or gemcitabine plus nab-paclitaxel chemotherapy). They reported that the number of CTC decreased or remained same in 12 (92.3%) patients with drug response (either tumor shrinkage or stable tumor burden). Negative enrichment, immunofluorescence, and in situ hybridization of chromosome 8 was applied to capturing CTC and determine the role of chromosomal instability in patients with PDAC. Improvement in detection rate of CTCs by using this system revealed the significance of triploid CTCs for the prediction of drug response to chemotherapy in these patients. In addition, CTMs correlated with poor response to chemotherapy in patients having stage IV PDAC[78].

The abundance of exosomal miRNAs was applied to assess therapeutic response. The expression levels of serum miRNAs (miR-221) was significantly upregulated at an earlier time, 3-6 wk of chemotherapy in patients with PDAC without drug response to lapatinib and capecitabine, compared to those without drug response. The expression levels of miRNA (specifically miR-221) increased in PDAC patients who did not respond to lapatinib and 5-fluorouracil (the active form of capecitabine), indicating that the increased level of specific serum miRNAs was associated with resistance to lapatinib and capecitabine treatment[136]. The levels of exosomes in serum at serial time points throughout chemoradiotherapy correlated with treatment resistance in 10 patients with locally advanced PDAC[137]. The levels of EphA2-EV in plasma were strongly associated with treatment response described in the early diagnosis section, reflecting the drug response to neoadjuvant therapy in 23 PDAC patients[84] (Table 4).

Table 4 Liquid biopsy in the therapeutic effect monitoring of pancreatic cancer.
Ref.
Journal
No. of patients
Biomarker
Method
Main findings
Del Re et al[133], 2017Sci Rep27ctDNAddPCRThere was a statistically significant difference in PFS and OS in patients with increase vs stability/reduction of ctDNA in the sample collected at day 15 (P = 0.03 and P = 0.009, respectively)
Kruger et al[134], 2018Ann Oncol54ctDNABEAMingAn increase in ctDNA at day 14 correlated with disease progression on subsequent imaging with a sensitivity of 83% and specificity of 100%
Watanabe et al[98], 2019PLoS One39ctDNAddPCRThe emergence of KRAS ctDNA in longitudinal tests was associated with prognosis (P < 0.005)
Wei et al[135], 2019Cancer Lett13 (chemotherapy group)CTCVimentin or EpCAM immobilized microfluidic chipIn patients exhibiting a response, their CTC counts decreased or remained the same, except for one case
Okubo et al[108], 2017Eur J Surg Oncol65CTCCellsearchThe overall survival rate was significantly lower in patients with than in those without CTCs even after chemotherapy and chemoradiotherapy (P = 0.045)
Xu et al[78], 2017Int J Mol Sci83CTCNE-iFISHThe proportion of triploid CTC detected by the NE-iFISH was significantly decreased after chemotherapy (P < 0.001)
Tian et al[136], 2016Oncol Lett17microRNART-qPCRSignificant upregulation of serum miRNAs (miR-21, miR-210, miR-221 and miR-7), at earlier time points (3-6 wk) was observed in non-responders of chemotherapy compared to responders
Bernard et al[99], 2019Gastroenterology104 (chemotherapy group)exosome and ctDNAddPCRIn the longitudinal analysis in chemotherapy group, a MAF peak above 1% in exosome DNA was significantly associated with radiologic progression (P = 0.0003)
An et al[137], 2017J Proteome Res10exosomeiTRAQThey analyzed exosomes before treatment, after one cycle of induction gemcitabine-based chemotherapy, and at 3 wk after starting chemoradiation therapy and compared these samples to serum derived from healthy volunteers. They identified eight proteins that changed during a course of therapy in all patients
Liang et al[84], 2017Nat Biomed Eng23 (neoadjuvant chemotherapy group)EVnPESEphA2-EVs were also informative in detecting early responses to neoadjuvant therapy (P < 0.05)
Yin et al[138], 2021Clin Cancer Res36somatic mutations, CTCs, and ctDNANext-generation sequencing & ISETSomatic mutations, CTCs, and ctDNA existed even in patients with PDAC with pathologic complete response to NAT, which could possibly predict early recurrence and reduced survival
Poruk et al[140], 2016Ann Surg50CTCISETThe detection of CTCs expressing both vimentin and cytokeratin was predictive of recurrence (P = 0.01)
Gemenetzis et al[139], 2018Ann Surg57CTCISETPatients who received neoadjuvant chemotherapy had significantly lower total CTCs (tCTCs, P = 0.007), eCTCs (P = 0.007), and mCTCs (P = 0.034), compared with untreated patients eligible for upfront resection
CONCLUSION

Liquid biopsy is gaining attention as a non-invasive methodology and is involved in obtaining important tumor information via blood-based biomarkers for early diagnosis and treatment of cancer. Tumor components such as CTCs, cfDNA, ctDNA, miRNAs, and exosomes in liquid biopsy have promising value for diagnosis, prognosis, and treatment prediction of surgery and chemotherapy. In particular, the most significant advantage of liquid biopsy over tissue biopsy is its ability to monitor disease progression and treatment efficacy longitudinally in "real time." However, liquid biopsy is not yet considered a standard means of confirming or diagnosing various diseases, including cancer.

The main limitation of liquid biopsy is its lack of sensitivity and accuracy in identifying various types of tumor compared to tissue biopsy. In addition, the ability to detect liquid biopsy is difficult because CTCs, ctDNA, and RNA are relatively scarce compared to other blood components. Furthermore, there are no standard separation, enrichment, and detection methodologies. Applying different techniques and assays to detect CTCs and ctDNA will result in varying sensitivity and specificity[141,142]. Usually, enriched CTCs are identified using tumor-associated biomarkers, either at the protein or mRNA level. However, in patients with epithelial tumors, epithelial markers are downregulated in the process of epithelial mesenchymal transition, making it difficult to identify them, leading to false-negative results[143]. Another limitation is the low specificity due to the presence of cfDNA from normal tissue. Furthermore, cfDNA is released from normal cells as a diluent for trace amounts of ctDNA, so an additional pre-step is needed in the analysis to avoid an increase in non-neoplastic cfDNA[144]. Establishing blood noncoding RNA as a biomarker is more complicated than in other common biomarkers due to its lack of suitable housekeeping noncoding RNA reference analytes, and high intra-patient variability. Thus, these limitations can lead to a lack of consistency between biomarkers identified in different studies[145]. Exosomes are more manageable to isolate than CTCs or cfDNA in tumors. Therefore, more studies focus on exosomes in the early diagnosis of cancer. However, they still have limitations in clinical application, such as low targeting efficiency and easy phagocytosis by the immune system. In addition, methods for isolating and purifying exosomes are time and labor-consuming. More multicentered, large-scale, and long-term studies, including clinical trials, are urgently needed to make liquid biopsy clinically available.

Similar to other tumors, cfDNA, ctDNA, CTCs, exosomes, and miRNAs are promising new biomarkers in the treatment of PDAC. However, the use of liquid biopsies in the same manner as conventional tumor markers of PDAC is not clear yet. These liquid biopsies are at least as effective as currently used tumor markers. Furthermore, cost-effectiveness is significant when the marker is used for clinical practice. With the advent of KRAS12c inhibitors, liquid biopsy will be performed many times during treatment. It is also possible that oncocytic carcinomas may acquire somatic mutations, such as EGFRT 790M in lung cancer, which must be monitored. Although liquid biopsy is minimally invasive, new and innovative technologies are needed to reduce the time and effort required for multiple analyses. With advances in cancer genomic medicine, new base mutation-specific inhibitors have been developed or new genetic mutations directly linked to drug resistance have been identified. So, strategies for various cancers may change at an unprecedented pace. Since it is practically difficult to make next-generation sequence analysis mandatory many times during treatment, it will be important in the future to assemble an appropriate analytical system that is as minimal as necessary.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Oncology

Country/Territory of origin: Japan

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Farolfi T, Italy; Yoon SB, South Korea S-Editor: Zhang H L-Editor: A P-Editor: Zhang H

References
1.  Tempero MA, Malafa MP, Al-Hawary M, Asbun H, Bain A, Behrman SW, Benson AB 3rd, Binder E, Cardin DB, Cha C, Chiorean EG, Chung V, Czito B, Dillhoff M, Dotan E, Ferrone CR, Hardacre J, Hawkins WG, Herman J, Ko AH, Komanduri S, Koong A, LoConte N, Lowy AM, Moravek C, Nakakura EK, O'Reilly EM, Obando J, Reddy S, Scaife C, Thayer S, Weekes CD, Wolff RA, Wolpin BM, Burns J, Darlow S. Pancreatic Adenocarcinoma, Version 2.2017, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2017;15:1028-1061.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 556]  [Cited by in F6Publishing: 668]  [Article Influence: 111.3]  [Reference Citation Analysis (0)]
2.  Silvestris N, Brunetti O, Bittoni A, Cataldo I, Corsi D, Crippa S, D'Onofrio M, Fiore M, Giommoni E, Milella M, Pezzilli R, Vasile E, Reni M. Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up of Exocrine Pancreatic Ductal Adenocarcinoma: Evidence Evaluation and Recommendations by the Italian Association of Medical Oncology (AIOM). Cancers (Basel). 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
3.  Winter K, Talar-Wojnarowska R, Dąbrowski A, Degowska M, Durlik M, Gąsiorowska A, Głuszek S, Jurkowska G, Kaczka A, Lampe P, Marek T, Nasierowska-Guttmejer A, Nowakowska-Duława E, Rydzewska G, Strzelczyk J, Śledziński Z, Małecka-Panas E. Diagnostic and therapeutic recommendations in pancreatic ductal adenocarcinoma. Recommendations of the Working Group of the Polish Pancreatic Club. Prz Gastroenterol. 2019;14:1-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
4.  Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin. 2013;63:11-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9215]  [Cited by in F6Publishing: 9738]  [Article Influence: 885.3]  [Reference Citation Analysis (3)]
5.  Hori M, Matsuda T, Shibata A, Katanoda K, Sobue T, Nishimoto H; Japan Cancer Surveillance Research Group. Cancer incidence and incidence rates in Japan in 2009: a study of 32 population-based cancer registries for the Monitoring of Cancer Incidence in Japan (MCIJ) project. Jpn J Clin Oncol. 2015;45:884-891.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 399]  [Cited by in F6Publishing: 438]  [Article Influence: 48.7]  [Reference Citation Analysis (0)]
6.  Gillen S, Schuster T, Meyer Zum Büschenfelde C, Friess H, Kleeff J. Preoperative/neoadjuvant therapy in pancreatic cancer: a systematic review and meta-analysis of response and resection percentages. PLoS Med. 2010;7:e1000267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1085]  [Cited by in F6Publishing: 1096]  [Article Influence: 78.3]  [Reference Citation Analysis (0)]
7.  Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74:2913-2921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3888]  [Cited by in F6Publishing: 4651]  [Article Influence: 465.1]  [Reference Citation Analysis (0)]
8.  Sivapalan L, Kocher HM, Ross-Adams H, Chelala C. Molecular profiling of ctDNA in pancreatic cancer: Opportunities and challenges for clinical application. Pancreatology. 2021;21:363-378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
9.  Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet. 2016;388:73-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1397]  [Cited by in F6Publishing: 1573]  [Article Influence: 196.6]  [Reference Citation Analysis (0)]
10.  Singh P, Srinivasan R, Wig JD. Major molecular markers in pancreatic ductal adenocarcinoma and their roles in screening, diagnosis, prognosis, and treatment. Pancreas. 2011;40:644-652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 43]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
11.  Barugola G, Partelli S, Marcucci S, Sartori N, Capelli P, Bassi C, Pederzoli P, Falconi M. Resectable pancreatic cancer: who really benefits from resection? Ann Surg Oncol. 2009;16:3316-3322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 121]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
12.  Hess V, Glimelius B, Grawe P, Dietrich D, Bodoky G, Ruhstaller T, Bajetta E, Saletti P, Figer A, Scheithauer W, Herrmann R. CA 19-9 tumour-marker response to chemotherapy in patients with advanced pancreatic cancer enrolled in a randomised controlled trial. Lancet Oncol. 2008;9:132-138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 145]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
13.  Humphris JL, Chang DK, Johns AL, Scarlett CJ, Pajic M, Jones MD, Colvin EK, Nagrial A, Chin VT, Chantrill LA, Samra JS, Gill AJ, Kench JG, Merrett ND, Das A, Musgrove EA, Sutherland RL, Biankin AV; NSW Pancreatic Cancer Network. The prognostic and predictive value of serum CA19.9 in pancreatic cancer. Ann Oncol. 2012;23:1713-1722.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 200]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
14.  Karachristos A, Scarmeas N, Hoffman JP. CA 19-9 levels predict results of staging laparoscopy in pancreatic cancer. J Gastrointest Surg. 2005;9:1286-1292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 76]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
15.  Maithel SK, Maloney S, Winston C, Gönen M, D'Angelica MI, Dematteo RP, Jarnagin WR, Brennan MF, Allen PJ. Preoperative CA 19-9 and the yield of staging laparoscopy in patients with radiographically resectable pancreatic adenocarcinoma. Ann Surg Oncol. 2008;15:3512-3520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 151]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
16.  Wasan HS, Springett GM, Chodkiewicz C, Wong R, Maurel J, Barone C, Rosbrook B, Ricart AD, Kim S, Spano JP. CA 19-9 as a biomarker in advanced pancreatic cancer patients randomised to gemcitabine plus axitinib or gemcitabine alone. Br J Cancer. 2009;101:1162-1167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 34]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
17.  Marchegiani G, Andrianello S, Malleo G, De Gregorio L, Scarpa A, Mino-Kenudson M, Maggino L, Ferrone CR, Lillemoe KD, Bassi C, Castillo CF, Salvia R. Does Size Matter in Pancreatic Cancer? Ann Surg. 2017;266:142-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 69]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
18.  Coppola A, La Vaccara V, Fiore M, Farolfi T, Ramella S, Angeletti S, Coppola R, Caputo D. CA19.9 Serum Level Predicts Lymph-Nodes Status in Resectable Pancreatic Ductal Adenocarcinoma: A Retrospective Single-Center Analysis. Front Oncol. 2021;11:690580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
19.  Ferrone CR, Finkelstein DM, Thayer SP, Muzikansky A, Fernandez-delCastillo C, Warshaw AL. Perioperative CA19-9 levels can predict stage and survival in patients with resectable pancreatic adenocarcinoma. J Clin Oncol. 2006;24:2897-2902.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 355]  [Cited by in F6Publishing: 393]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
20.  Dong Q, Yang XH, Zhang Y, Jing W, Zheng LQ, Liu YP, Qu XJ. Elevated serum CA19-9 level is a promising predictor for poor prognosis in patients with resectable pancreatic ductal adenocarcinoma: a pilot study. World J Surg Oncol. 2014;12:171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 42]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
21.  Imaoka H, Shimizu Y, Senda Y, Natsume S, Mizuno N, Hara K, Hijioka S, Hieda N, Tajika M, Tanaka T, Ishihara M, Niwa Y, Yamao K. Post-adjuvant chemotherapy CA19-9 levels predict prognosis in patients with pancreatic ductal adenocarcinoma: A retrospective cohort study. Pancreatology. 2016;16:658-664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
22.  Laurent L, Sefrioui D, Bignon AL, Parzy A, Sidali S, Hassine M, Gangloff A, Galais MP, Bouhier-Leporrier K, Michel P, Di Fiore F. CA19.9 decrease >15% is a predictor of favourable outcome in patients treated for advanced pancreatic carcinoma: analysis of two independent cohorts. HPB (Oxford). 2019;21:582-588.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
23.  Robert M, Jarlier M, Gourgou S, Desseigne F, Ychou M, Bouché O, Juzyna B, Conroy T, Bennouna J. Retrospective Analysis of CA19-9 Decrease in Patients with Metastatic Pancreatic Carcinoma Treated with FOLFIRINOX or Gemcitabine in a Randomized Phase III Study (ACCORD11/PRODIGE4). Oncology. 2017;93:367-376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 42]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
24.  Tsai S, George B, Wittmann D, Ritch PS, Krepline AN, Aldakkak M, Barnes CA, Christians KK, Dua K, Griffin M, Hagen C, Hall WA, Erickson BA, Evans DB. Importance of Normalization of CA19-9 Levels Following Neoadjuvant Therapy in Patients With Localized Pancreatic Cancer. Ann Surg. 2020;271:740-747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 109]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
25.  Montgomery RC, Hoffman JP, Riley LB, Rogatko A, Ridge JA, Eisenberg BL. Prediction of recurrence and survival by post-resection CA 19-9 values in patients with adenocarcinoma of the pancreas. Ann Surg Oncol. 1997;4:551-556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 102]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
26.  Nakao A, Oshima K, Nomoto S, Takeda S, Kaneko T, Ichihara T, Kurokawa T, Nonami T, Takagi H. Clinical usefulness of CA-19-9 in pancreatic carcinoma. Semin Surg Oncol. 1998;15:15-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
27.  Luo G, Fan Z, Cheng H, Jin K, Guo M, Lu Y, Yang C, Fan K, Huang Q, Long J, Liu L, Xu J, Lu R, Ni Q, Warshaw AL, Liu C, Yu X. New observations on the utility of CA19-9 as a biomarker in Lewis negative patients with pancreatic cancer. Pancreatology. 2018;18:971-976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 38]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
28.  Joosse SA, Pantel K. Tumor-Educated Platelets as Liquid Biopsy in Cancer Patients. Cancer Cell. 2015;28:552-554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 107]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
29.  Bagcchi S. Urine test can detect early stage pancreatic cancer. Lancet Oncol. 2015;16:e431.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 5]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
30.  Adams DL, Martin SS, Alpaugh RK, Charpentier M, Tsai S, Bergan RC, Ogden IM, Catalona W, Chumsri S, Tang CM, Cristofanilli M. Circulating giant macrophages as a potential biomarker of solid tumors. Proc Natl Acad Sci U S A. 2014;111:3514-3519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 192]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
31.  Schwarzenbach H, Nishida N, Calin GA, Pantel K. Clinical relevance of circulating cell-free microRNAs in cancer. Nat Rev Clin Oncol. 2014;11:145-156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 704]  [Cited by in F6Publishing: 789]  [Article Influence: 78.9]  [Reference Citation Analysis (0)]
32.  Best MG, Sol N, Kooi I, Tannous J, Westerman BA, Rustenburg F, Schellen P, Verschueren H, Post E, Koster J, Ylstra B, Ameziane N, Dorsman J, Smit EF, Verheul HM, Noske DP, Reijneveld JC, Nilsson RJA, Tannous BA, Wesseling P, Wurdinger T. RNA-Seq of Tumor-Educated Platelets Enables Blood-Based Pan-Cancer, Multiclass, and Molecular Pathway Cancer Diagnostics. Cancer Cell. 2015;28:666-676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 569]  [Cited by in F6Publishing: 555]  [Article Influence: 61.7]  [Reference Citation Analysis (0)]
33.  Melo SA, Luecke LB, Kahlert C, Fernandez AF, Gammon ST, Kaye J, LeBleu VS, Mittendorf EA, Weitz J, Rahbari N, Reissfelder C, Pilarsky C, Fraga MF, Piwnica-Worms D, Kalluri R. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature. 2015;523:177-182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2108]  [Cited by in F6Publishing: 1977]  [Article Influence: 219.7]  [Reference Citation Analysis (0)]
34.  Lui YY, Chik KW, Chiu RW, Ho CY, Lam CW, Lo YM. Predominant hematopoietic origin of cell-free DNA in plasma and serum after sex-mismatched bone marrow transplantation. Clin Chem. 2002;48:421-427.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Sun K, Jiang P, Chan KC, Wong J, Cheng YK, Liang RH, Chan WK, Ma ES, Chan SL, Cheng SH, Chan RW, Tong YK, Ng SS, Wong RS, Hui DS, Leung TN, Leung TY, Lai PB, Chiu RW, Lo YM. Plasma DNA tissue mapping by genome-wide methylation sequencing for noninvasive prenatal, cancer, and transplantation assessments. Proc Natl Acad Sci U S A. 2015;112:E5503-E5512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 416]  [Cited by in F6Publishing: 460]  [Article Influence: 51.1]  [Reference Citation Analysis (0)]
36.  Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 1977;37:646-650.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Lo YM, Corbetta N, Chamberlain PF, Rai V, Sargent IL, Redman CW, Wainscoat JS. Presence of fetal DNA in maternal plasma and serum. Lancet. 1997;350:485-487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2104]  [Cited by in F6Publishing: 1944]  [Article Influence: 72.0]  [Reference Citation Analysis (0)]
38.  Lo YM, Tein MS, Pang CC, Yeung CK, Tong KL, Hjelm NM. Presence of donor-specific DNA in plasma of kidney and liver-transplant recipients. Lancet. 1998;351:1329-1330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 227]  [Cited by in F6Publishing: 217]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
39.  Mandel P, Metais P. Nuclear Acids In Human Blood Plasma. C R Seances Soc Biol Fil. 1948;142:241-243.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Stroun M, Anker P, Maurice P, Lyautey J, Lederrey C, Beljanski M. Neoplastic characteristics of the DNA found in the plasma of cancer patients. Oncology. 1989;46:318-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 474]  [Cited by in F6Publishing: 485]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
41.  Underhill HR, Kitzman JO, Hellwig S, Welker NC, Daza R, Baker DN, Gligorich KM, Rostomily RC, Bronner MP, Shendure J. Fragment Length of Circulating Tumor DNA. PLoS Genet. 2016;12:e1006162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 374]  [Cited by in F6Publishing: 435]  [Article Influence: 54.4]  [Reference Citation Analysis (0)]
42.  Pierga JY, Bidard FC, Mathiot C, Brain E, Delaloge S, Giachetti S, de Cremoux P, Salmon R, Vincent-Salomon A, Marty M. Circulating tumor cell detection predicts early metastatic relapse after neoadjuvant chemotherapy in large operable and locally advanced breast cancer in a phase II randomized trial. Clin Cancer Res. 2008;14:7004-7010.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 295]  [Cited by in F6Publishing: 287]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
43.  Camara O, Rengsberger M, Egbe A, Koch A, Gajda M, Hammer U, Jörke C, Rabenstein C, Untch M, Pachmann K. The relevance of circulating epithelial tumor cells (CETC) for therapy monitoring during neoadjuvant (primary systemic) chemotherapy in breast cancer. Ann Oncol. 2007;18:1484-1492.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 78]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
44.  de Bono JS, Scher HI, Montgomery RB, Parker C, Miller MC, Tissing H, Doyle GV, Terstappen LW, Pienta KJ, Raghavan D. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin Cancer Res. 2008;14:6302-6309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1588]  [Cited by in F6Publishing: 1646]  [Article Influence: 102.9]  [Reference Citation Analysis (0)]
45.  Yousefi M, Ghaffari P, Nosrati R, Dehghani S, Salmaninejad A, Abarghan YJ, Ghaffari SH. Prognostic and therapeutic significance of circulating tumor cells in patients with lung cancer. Cell Oncol (Dordr). 2020;43:31-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
46.  Cohen SJ, Punt CJ, Iannotti N, Saidman BH, Sabbath KD, Gabrail NY, Picus J, Morse MA, Mitchell E, Miller MC, Doyle GV, Tissing H, Terstappen LW, Meropol NJ. Prognostic significance of circulating tumor cells in patients with metastatic colorectal cancer. Ann Oncol. 2009;20:1223-1229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 370]  [Cited by in F6Publishing: 381]  [Article Influence: 25.4]  [Reference Citation Analysis (0)]
47.  Yu M, Stott S, Toner M, Maheswaran S, Haber DA. Circulating tumor cells: approaches to isolation and characterization. J Cell Biol. 2011;192:373-382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 820]  [Cited by in F6Publishing: 784]  [Article Influence: 60.3]  [Reference Citation Analysis (0)]
48.  Aceto N, Bardia A, Miyamoto DT, Donaldson MC, Wittner BS, Spencer JA, Yu M, Pely A, Engstrom A, Zhu H, Brannigan BW, Kapur R, Stott SL, Shioda T, Ramaswamy S, Ting DT, Lin CP, Toner M, Haber DA, Maheswaran S. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell. 2014;158:1110-1122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1463]  [Cited by in F6Publishing: 1610]  [Article Influence: 178.9]  [Reference Citation Analysis (0)]
49.  Murlidhar V, Reddy RM, Fouladdel S, Zhao L, Ishikawa MK, Grabauskiene S, Zhang Z, Lin J, Chang AC, Carrott P, Lynch WR, Orringer MB, Kumar-Sinha C, Palanisamy N, Beer DG, Wicha MS, Ramnath N, Azizi E, Nagrath S. Poor Prognosis Indicated by Venous Circulating Tumor Cell Clusters in Early-Stage Lung Cancers. Cancer Res. 2017;77:5194-5206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 126]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
50.  Fidler IJ. The relationship of embolic homogeneity, number, size and viability to the incidence of experimental metastasis. Eur J Cancer (1965). 1973;9:223-227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 282]  [Cited by in F6Publishing: 259]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
51.  Liotta LA, Saidel MG, Kleinerman J. The significance of hematogenous tumor cell clumps in the metastatic process. Cancer Res. 1976;36:889-894.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Mu Z, Wang C, Ye Z, Austin L, Civan J, Hyslop T, Palazzo JP, Jaslow R, Li B, Myers RE, Jiang J, Xing J, Yang H, Cristofanilli M. Prospective assessment of the prognostic value of circulating tumor cells and their clusters in patients with advanced-stage breast cancer. Breast Cancer Res Treat. 2015;154:563-571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 93]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
53.  Molnar B, Floro L, Sipos F, Toth B, Sreter L, Tulassay Z. Elevation in peripheral blood circulating tumor cell number correlates with macroscopic progression in UICC stage IV colorectal cancer patients. Dis Markers. 2008;24:141-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 24]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
54.  Wolf P. The nature and significance of platelet products in human plasma. Br J Haematol. 1967;13:269-288.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1035]  [Cited by in F6Publishing: 1104]  [Article Influence: 19.4]  [Reference Citation Analysis (1)]
55.  Pan BT, Johnstone RM. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Cell. 1983;33:967-978.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1188]  [Cited by in F6Publishing: 1320]  [Article Influence: 32.2]  [Reference Citation Analysis (0)]
56.  Théry C. Exosomes: secreted vesicles and intercellular communications. F1000 Biol Rep. 2011;3:15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 583]  [Cited by in F6Publishing: 681]  [Article Influence: 52.4]  [Reference Citation Analysis (0)]
57.  Kalluri R. The biology and function of exosomes in cancer. J Clin Invest. 2016;126:1208-1215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 944]  [Cited by in F6Publishing: 1231]  [Article Influence: 153.9]  [Reference Citation Analysis (0)]
58.  Vader P, Breakefield XO, Wood MJ. Extracellular vesicles: emerging targets for cancer therapy. Trends Mol Med. 2014;20:385-393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 283]  [Cited by in F6Publishing: 304]  [Article Influence: 30.4]  [Reference Citation Analysis (0)]
59.  Skog J, Würdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, Curry WT Jr, Carter BS, Krichevsky AM, Breakefield XO. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10:1470-1476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3409]  [Cited by in F6Publishing: 3723]  [Article Influence: 232.7]  [Reference Citation Analysis (0)]
60.  Raposo G, Nijman HW, Stoorvogel W, Liejendekker R, Harding CV, Melief CJ, Geuze HJ. B lymphocytes secrete antigen-presenting vesicles. J Exp Med. 1996;183:1161-1172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2293]  [Cited by in F6Publishing: 2484]  [Article Influence: 88.7]  [Reference Citation Analysis (0)]
61.  Zitvogel L, Regnault A, Lozier A, Wolfers J, Flament C, Tenza D, Ricciardi-Castagnoli P, Raposo G, Amigorena S. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med. 1998;4:594-600.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1514]  [Cited by in F6Publishing: 1578]  [Article Influence: 60.7]  [Reference Citation Analysis (0)]
62.  Meehan K, Vella LJ. The contribution of tumour-derived exosomes to the hallmarks of cancer. Crit Rev Clin Lab Sci. 2016;53:121-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 91]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
63.  Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75:843-854.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8672]  [Cited by in F6Publishing: 8451]  [Article Influence: 272.6]  [Reference Citation Analysis (0)]
64.  Tüfekci KU, Oner MG, Meuwissen RL, Genç S. The role of microRNAs in human diseases. Methods Mol Biol. 2014;1107:33-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 159]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
65.  Gasparri ML, Casorelli A, Bardhi E, Besharat AR, Savone D, Ruscito I, Farooqi AA, Papadia A, Mueller MD, Ferretti E, Benedetti Panici P. Beyond circulating microRNA biomarkers: Urinary microRNAs in ovarian and breast cancer. Tumour Biol. 2017;39:1010428317695525.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
66.  Akao Y, Nakagawa Y, Hirata I, Iio A, Itoh T, Kojima K, Nakashima R, Kitade Y, Naoe T. Role of anti-oncomirs miR-143 and -145 in human colorectal tumors. Cancer Gene Ther. 2010;17:398-408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 197]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
67.  Montagnana M, Benati M, Danese E, Giudici S, Perfranceschi M, Ruzzenenete O, Salvagno GL, Bassi A, Gelati M, Paviati E, Guidi GC, Franchi M, Lippi G. Aberrant MicroRNA Expression in Patients With Endometrial Cancer. Int J Gynecol Cancer. 2017;27:459-466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
68.  Eismann J, Hirschfeld M, Erbes T, Rücker G, Jäger M, Ritter A, Weiss D, Gitsch G, Mayer S. Hypoxia- and acidosis-driven aberrations of secreted microRNAs in endometrial cancer in vitro. Oncol Rep. 2017;38:993-1004.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
69.  Szymanski M, Barciszewska MZ, Erdmann VA, Barciszewski J. A new frontier for molecular medicine: noncoding RNAs. Biochim Biophys Acta. 2005;1756:65-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 46]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
70.  Bardeesy N, DePinho RA. Pancreatic cancer biology and genetics. Nat Rev Cancer. 2002;2:897-909.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 848]  [Cited by in F6Publishing: 826]  [Article Influence: 37.5]  [Reference Citation Analysis (0)]
71.  Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, Mollaee M, Wagner KU, Koduru P, Yopp A, Choti MA, Yeo CJ, McCue P, White MA, Knudsen ES. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun. 2015;6:6744.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 704]  [Cited by in F6Publishing: 764]  [Article Influence: 84.9]  [Reference Citation Analysis (0)]
72.  Hashimoto D, Arima K, Yokoyama N, Chikamoto A, Taki K, Inoue R, Kaida T, Higashi T, Nitta H, Ohmuraya M, Hirota M, Beppu T, Baba H. Heterogeneity of KRAS Mutations in Pancreatic Ductal Adenocarcinoma. Pancreas. 2016;45:1111-1114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
73.  Makohon-Moore AP, Zhang M, Reiter JG, Bozic I, Allen B, Kundu D, Chatterjee K, Wong F, Jiao Y, Kohutek ZA, Hong J, Attiyeh M, Javier B, Wood LD, Hruban RH, Nowak MA, Papadopoulos N, Kinzler KW, Vogelstein B, Iacobuzio-Donahue CA. Limited heterogeneity of known driver gene mutations among the metastases of individual patients with pancreatic cancer. Nat Genet. 2017;49:358-366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 250]  [Cited by in F6Publishing: 263]  [Article Influence: 37.6]  [Reference Citation Analysis (0)]
74.  Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, Thornton K, Agrawal N, Sokoll L, Szabo SA, Kinzler KW, Vogelstein B, Diaz LA Jr. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14:985-990.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1719]  [Cited by in F6Publishing: 1909]  [Article Influence: 112.3]  [Reference Citation Analysis (0)]
75.  Chused TM, Steinberg AD, Talal N. The clearance and localization of nucleic acids by New Zealand and normal mice. Clin Exp Immunol. 1972;12:465-476.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Ankeny JS, Court CM, Hou S, Li Q, Song M, Wu D, Chen JF, Lee T, Lin M, Sho S, Rochefort MM, Girgis MD, Yao J, Wainberg ZA, Muthusamy VR, Watson RR, Donahue TR, Hines OJ, Reber HA, Graeber TG, Tseng HR, Tomlinson JS. Circulating tumour cells as a biomarker for diagnosis and staging in pancreatic cancer. Br J Cancer. 2016;114:1367-1375.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 103]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
77.  Rhim AD, Thege FI, Santana SM, Lannin TB, Saha TN, Tsai S, Maggs LR, Kochman ML, Ginsberg GG, Lieb JG, Chandrasekhara V, Drebin JA, Ahmad N, Yang YX, Kirby BJ, Stanger BZ. Detection of circulating pancreas epithelial cells in patients with pancreatic cystic lesions. Gastroenterology. 2014;146:647-651.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 173]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
78.  Xu Y, Qin T, Li J, Wang X, Gao C, Xu C, Hao J, Liu J, Gao S, Ren H. Detection of Circulating Tumor Cells Using Negative Enrichment Immunofluorescence and an In Situ Hybridization System in Pancreatic Cancer. Int J Mol Sci. 2017;18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 34]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
79.  Poruk KE, Blackford AL, Weiss MJ, Cameron JL, He J, Goggins M, Rasheed ZA, Wolfgang CL, Wood LD. Circulating Tumor Cells Expressing Markers of Tumor-Initiating Cells Predict Poor Survival and Cancer Recurrence in Patients with Pancreatic Ductal Adenocarcinoma. Clin Cancer Res. 2017;23:2681-2690.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 79]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
80.  Gao Y, Zhu Y, Zhang Z, Zhang C, Huang X, Yuan Z. Clinical significance of pancreatic circulating tumor cells using combined negative enrichment and immunostaining-fluorescence in situ hybridization. J Exp Clin Cancer Res. 2016;35:66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 63]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
81.  Kulemann B, Pitman MB, Liss AS, Valsangkar N, Fernández-Del Castillo C, Lillemoe KD, Hoeppner J, Mino-Kenudson M, Warshaw AL, Thayer SP. Circulating tumor cells found in patients with localized and advanced pancreatic cancer. Pancreas. 2015;44:547-550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 74]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
82.  Buscail E, Alix-Panabières C, Quincy P, Cauvin T, Chauvet A, Degrandi O, Caumont C, Verdon S, Lamrissi I, Moranvillier I, Buscail C, Marty M, Laurent C, Vendrely V, Moreau-Gaudry F, Bedel A, Dabernat S, Chiche L. High Clinical Value of Liquid Biopsy to Detect Circulating Tumor Cells and Tumor Exosomes in Pancreatic Ductal Adenocarcinoma Patients Eligible for Up-Front Surgery. Cancers (Basel). 2019;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 73]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
83.  Lai X, Wang M, McElyea SD, Sherman S, House M, Korc M. A microRNA signature in circulating exosomes is superior to exosomal glypican-1 levels for diagnosing pancreatic cancer. Cancer Lett. 2017;393:86-93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 234]  [Article Influence: 33.4]  [Reference Citation Analysis (0)]
84.  Liang K, Liu F, Fan J, Sun D, Liu C, Lyon CJ, Bernard DW, Li Y, Yokoi K, Katz MH, Koay EJ, Zhao Z, Hu Y. Nanoplasmonic Quantification of Tumor-derived Extracellular Vesicles in Plasma Microsamples for Diagnosis and Treatment Monitoring. Nat Biomed Eng. 2017;1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 208]  [Cited by in F6Publishing: 246]  [Article Influence: 35.1]  [Reference Citation Analysis (0)]
85.  Que R, Ding G, Chen J, Cao L. Analysis of serum exosomal microRNAs and clinicopathologic features of patients with pancreatic adenocarcinoma. World J Surg Oncol. 2013;11:219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 198]  [Article Influence: 18.0]  [Reference Citation Analysis (1)]
86.  Cote GA, Gore AJ, McElyea SD, Heathers LE, Xu H, Sherman S, Korc M. A pilot study to develop a diagnostic test for pancreatic ductal adenocarcinoma based on differential expression of select miRNA in plasma and bile. Am J Gastroenterol. 2014;109:1942-1952.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 86]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
87.  Ouyang H, Gore J, Deitz S, Korc M. microRNA-10b enhances pancreatic cancer cell invasion by suppressing TIP30 expression and promoting EGF and TGF-β actions. Oncogene. 2014;33:4664-4674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 94]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
88.  Slater EP, Strauch K, Rospleszcz S, Ramaswamy A, Esposito I, Klöppel G, Matthäi E, Heeger K, Fendrich V, Langer P, Bartsch DK. MicroRNA-196a and -196b as Potential Biomarkers for the Early Detection of Familial Pancreatic Cancer. Transl Oncol. 2014;7:464-471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 58]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
89.  Madhavan B, Yue S, Galli U, Rana S, Gross W, Müller M, Giese NA, Kalthoff H, Becker T, Büchler MW, Zöller M. Combined evaluation of a panel of protein and miRNA serum-exosome biomarkers for pancreatic cancer diagnosis increases sensitivity and specificity. Int J Cancer. 2015;136:2616-2627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 314]  [Cited by in F6Publishing: 358]  [Article Influence: 35.8]  [Reference Citation Analysis (0)]
90.  Peng C, Wang J, Gao W, Huang L, Liu Y, Li X, Li Z, Yu X. Meta-analysis of the Diagnostic Performance of Circulating MicroRNAs for Pancreatic Cancer. Int J Med Sci. 2021;18:660-671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
91.  Singh N, Gupta S, Pandey RM, Chauhan SS, Saraya A. High levels of cell-free circulating nucleic acids in pancreatic cancer are associated with vascular encasement, metastasis and poor survival. Cancer Invest. 2015;33:78-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 68]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
92.  Lapin M, Oltedal S, Tjensvoll K, Buhl T, Smaaland R, Garresori H, Javle M, Glenjen NI, Abelseth BK, Gilje B, Nordgård O. Fragment size and level of cell-free DNA provide prognostic information in patients with advanced pancreatic cancer. J Transl Med. 2018;16:300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 66]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
93.  Wang SE, Shyr BU, Shyr BS, Chen SC, Chang SC, Shyr YM. Circulating Cell-Free DNA in Pancreatic Head Adenocarcinoma Undergoing Pancreaticoduodenectomy. Pancreas. 2021;50:214-218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
94.  Castells A, Puig P, Móra J, Boadas J, Boix L, Urgell E, Solé M, Capellà G, Lluís F, Fernández-Cruz L, Navarro S, Farré A. K-ras mutations in DNA extracted from the plasma of patients with pancreatic carcinoma: diagnostic utility and prognostic significance. J Clin Oncol. 1999;17:578-584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 172]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
95.  Ako S, Nouso K, Kinugasa H, Dohi C, Matushita H, Mizukawa S, Muro S, Akimoto Y, Uchida D, Tomoda T, Matsumoto K, Horiguchi S, Tsutsumi K, Kato H, Okada H. Utility of serum DNA as a marker for KRAS mutations in pancreatic cancer tissue. Pancreatology. 2017;17:285-290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
96.  Hadano N, Murakami Y, Uemura K, Hashimoto Y, Kondo N, Nakagawa N, Sueda T, Hiyama E. Prognostic value of circulating tumour DNA in patients undergoing curative resection for pancreatic cancer. Br J Cancer. 2016;115:59-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 113]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
97.  Nakano Y, Kitago M, Matsuda S, Nakamura Y, Fujita Y, Imai S, Shinoda M, Yagi H, Abe Y, Hibi T, Fujii-Nishimura Y, Takeuchi A, Endo Y, Itano O, Kitagawa Y. KRAS mutations in cell-free DNA from preoperative and postoperative sera as a pancreatic cancer marker: a retrospective study. Br J Cancer. 2018;118:662-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 60]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
98.  Watanabe F, Suzuki K, Tamaki S, Abe I, Endo Y, Takayama Y, Ishikawa H, Kakizawa N, Saito M, Futsuhara K, Noda H, Konishi F, Rikiyama T. Longitudinal monitoring of KRAS-mutated circulating tumor DNA enables the prediction of prognosis and therapeutic responses in patients with pancreatic cancer. PLoS One. 2019;14:e0227366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 50]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
99.  Bernard V, Kim DU, San Lucas FA, Castillo J, Allenson K, Mulu FC, Stephens BM, Huang J, Semaan A, Guerrero PA, Kamyabi N, Zhao J, Hurd MW, Koay EJ, Taniguchi CM, Herman JM, Javle M, Wolff R, Katz M, Varadhachary G, Maitra A, Alvarez HA. Circulating Nucleic Acids Are Associated With Outcomes of Patients With Pancreatic Cancer. Gastroenterology. 2019;156:108-118.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 235]  [Article Influence: 47.0]  [Reference Citation Analysis (0)]
100.  Kinugasa H, Nouso K, Miyahara K, Morimoto Y, Dohi C, Tsutsumi K, Kato H, Matsubara T, Okada H, Yamamoto K. Detection of K-ras gene mutation by liquid biopsy in patients with pancreatic cancer. Cancer. 2015;121:2271-2280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 172]  [Article Influence: 19.1]  [Reference Citation Analysis (0)]
101.  Tjensvoll K, Lapin M, Buhl T, Oltedal S, Steen-Ottosen Berry K, Gilje B, Søreide JA, Javle M, Nordgård O, Smaaland R. Clinical relevance of circulating KRAS mutated DNA in plasma from patients with advanced pancreatic cancer. Mol Oncol. 2016;10:635-643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 111]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
102.  Chen H, Tu H, Meng ZQ, Chen Z, Wang P, Liu LM. K-ras mutational status predicts poor prognosis in unresectable pancreatic cancer. Eur J Surg Oncol. 2010;36:657-662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 97]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
103.  Sausen M, Phallen J, Adleff V, Jones S, Leary RJ, Barrett MT, Anagnostou V, Parpart-Li S, Murphy D, Kay Li Q, Hruban CA, Scharpf R, White JR, O'Dwyer PJ, Allen PJ, Eshleman JR, Thompson CB, Klimstra DS, Linehan DC, Maitra A, Hruban RH, Diaz LA Jr, Von Hoff DD, Johansen JS, Drebin JA, Velculescu VE. Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients. Nat Commun. 2015;6:7686.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 295]  [Cited by in F6Publishing: 322]  [Article Influence: 35.8]  [Reference Citation Analysis (0)]
104.  Han L, Chen W, Zhao Q. Prognostic value of circulating tumor cells in patients with pancreatic cancer: a meta-analysis. Tumour Biol. 2014;35:2473-2480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 82]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
105.  Khoja L, Backen A, Sloane R, Menasce L, Ryder D, Krebs M, Board R, Clack G, Hughes A, Blackhall F, Valle JW, Dive C. A pilot study to explore circulating tumour cells in pancreatic cancer as a novel biomarker. Br J Cancer. 2012;106:508-516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 201]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
106.  Earl J, Garcia-Nieto S, Martinez-Avila JC, Montans J, Sanjuanbenito A, Rodríguez-Garrote M, Lisa E, Mendía E, Lobo E, Malats N, Carrato A, Guillen-Ponce C. Circulating tumor cells (Ctc) and kras mutant circulating free Dna (cfdna) detection in peripheral blood as biomarkers in patients diagnosed with exocrine pancreatic cancer. BMC Cancer. 2015;15:797.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 132]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
107.  Zhang Y, Wang F, Ning N, Chen Q, Yang Z, Guo Y, Xu D, Zhang D, Zhan T, Cui W. Patterns of circulating tumor cells identified by CEP8, CK and CD45 in pancreatic cancer. Int J Cancer. 2015;136:1228-1233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 71]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
108.  Okubo K, Uenosono Y, Arigami T, Mataki Y, Matsushita D, Yanagita S, Kurahara H, Sakoda M, Kijima Y, Maemura K, Natsugoe S. Clinical impact of circulating tumor cells and therapy response in pancreatic cancer. Eur J Surg Oncol. 2017;43:1050-1055.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 57]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
109.  Chang MC, Chang YT, Chen JY, Jeng YM, Yang CY, Tien YW, Yang SH, Chen HL, Liang TY, Wang CF, Lee EY, Chang YC, Lee WH. Clinical Significance of Circulating Tumor Microemboli as a Prognostic Marker in Patients with Pancreatic Ductal Adenocarcinoma. Clin Chem. 2016;62:505-513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 66]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
110.  Bidard FC, Huguet F, Louvet C, Mineur L, Bouché O, Chibaudel B, Artru P, Desseigne F, Bachet JB, Mathiot C, Pierga JY, Hammel P. Circulating tumor cells in locally advanced pancreatic adenocarcinoma: the ancillary CirCe 07 study to the LAP 07 trial. Ann Oncol. 2013;24:2057-2061.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 141]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
111.  Kurihara T, Itoi T, Sofuni A, Itokawa F, Tsuchiya T, Tsuji S, Ishii K, Ikeuchi N, Tsuchida A, Kasuya K, Kawai T, Sakai Y, Moriyasu F. Detection of circulating tumor cells in patients with pancreatic cancer: a preliminary result. J Hepatobiliary Pancreat Surg. 2008;15:189-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 118]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
112.  de Albuquerque A, Kubisch I, Breier G, Stamminger G, Fersis N, Eichler A, Kaul S, Stölzel U. Multimarker gene analysis of circulating tumor cells in pancreatic cancer patients: a feasibility study. Oncology. 2012;82:3-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 89]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
113.  Kulemann B, Liss AS, Warshaw AL, Seifert S, Bronsert P, Glatz T, Pitman MB, Hoeppner J. KRAS mutations in pancreatic circulating tumor cells: a pilot study. Tumour Biol. 2016;37:7547-7554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 36]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
114.  Thind A, Wilson C. Exosomal miRNAs as cancer biomarkers and therapeutic targets. J Extracell Vesicles. 2016;5:31292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 294]  [Cited by in F6Publishing: 271]  [Article Influence: 33.9]  [Reference Citation Analysis (0)]
115.  Pitt JM, Kroemer G, Zitvogel L. Extracellular vesicles: masters of intercellular communication and potential clinical interventions. J Clin Invest. 2016;126:1139-1143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 271]  [Cited by in F6Publishing: 335]  [Article Influence: 41.9]  [Reference Citation Analysis (0)]
116.  Li Z, Tao Y, Wang X, Jiang P, Li J, Peng M, Zhang X, Chen K, Liu H, Zhen P, Zhu J, Liu X. Tumor-Secreted Exosomal miR-222 Promotes Tumor Progression via Regulating P27 Expression and Re-Localization in Pancreatic Cancer. Cell Physiol Biochem. 2018;51:610-629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 89]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
117.  Wang X, Luo G, Zhang K, Cao J, Huang C, Jiang T, Liu B, Su L, Qiu Z. Hypoxic Tumor-Derived Exosomal miR-301a Mediates M2 Macrophage Polarization via PTEN/PI3Kγ to Promote Pancreatic Cancer Metastasis. Cancer Res. 2018;78:4586-4598.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 420]  [Cited by in F6Publishing: 440]  [Article Influence: 73.3]  [Reference Citation Analysis (0)]
118.  Frampton AE, Prado MM, López-Jiménez E, Fajardo-Puerta AB, Jawad ZAR, Lawton P, Giovannetti E, Habib NA, Castellano L, Stebbing J, Krell J, Jiao LR. Glypican-1 is enriched in circulating-exosomes in pancreatic cancer and correlates with tumor burden. Oncotarget. 2018;9:19006-19013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 103]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
119.  Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, Becker A, Hoshino A, Mark MT, Molina H, Xiang J, Zhang T, Theilen TM, García-Santos G, Williams C, Ararso Y, Huang Y, Rodrigues G, Shen TL, Labori KJ, Lothe IM, Kure EH, Hernandez J, Doussot A, Ebbesen SH, Grandgenett PM, Hollingsworth MA, Jain M, Mallya K, Batra SK, Jarnagin WR, Schwartz RE, Matei I, Peinado H, Stanger BZ, Bromberg J, Lyden D. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. 2015;17:816-826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1554]  [Cited by in F6Publishing: 1849]  [Article Influence: 205.4]  [Reference Citation Analysis (0)]
120.  Hussung S, Akhoundova D, Hipp J, Follo M, Klar RFU, Philipp U, Scherer F, von Bubnoff N, Duyster J, Boerries M, Wittel U, Fritsch RM. Longitudinal analysis of cell-free mutated KRAS and CA 19-9 predicts survival following curative resection of pancreatic cancer. BMC Cancer. 2021;21:49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
121.  Groot VP, Mosier S, Javed AA, Teinor JA, Gemenetzis G, Ding D, Haley LM, Yu J, Burkhart RA, Hasanain A, Debeljak M, Kamiyama H, Narang A, Laheru DA, Zheng L, Lin MT, Gocke CD, Fishman EK, Hruban RH, Goggins MG, Molenaar IQ, Cameron JL, Weiss MJ, Velculescu VE, He J, Wolfgang CL, Eshleman JR. Circulating Tumor DNA as a Clinical Test in Resected Pancreatic Cancer. Clin Cancer Res. 2019;25:4973-4984.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 102]  [Article Influence: 20.4]  [Reference Citation Analysis (0)]
122.  Jiang J, Ye S, Xu Y, Chang L, Hu X, Ru G, Guo Y, Yi X, Yang L, Huang D. Circulating Tumor DNA as a Potential Marker to Detect Minimal Residual Disease and Predict Recurrence in Pancreatic Cancer. Front Oncol. 2020;10:1220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 27]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
123.  Kim MK, Woo SM, Park B, Yoon KA, Kim YH, Joo J, Lee WJ, Han SS, Park SJ, Kong SY. Prognostic Implications of Multiplex Detection of KRAS Mutations in Cell-Free DNA from Patients with Pancreatic Ductal Adenocarcinoma. Clin Chem. 2018;64:726-734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 61]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
124.  Yamaguchi T, Uemura K, Murakami Y, Kondo N, Nakagawa N, Okada K, Seo S, Hiyama E, Takahashi S, Sueda T. Clinical Implications of Pre- and Postoperative Circulating Tumor DNA in Patients with Resected Pancreatic Ductal Adenocarcinoma. Ann Surg Oncol. 2021;28:3135-3144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
125.  Guo S, Shi X, Shen J, Gao S, Wang H, Shen S, Pan Y, Li B, Xu X, Shao Z, Jin G. Preoperative detection of KRAS G12D mutation in ctDNA is a powerful predictor for early recurrence of resectable PDAC patients. Br J Cancer. 2020;122:857-867.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 43]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
126.  Lee B, Lipton L, Cohen J, Tie J, Javed AA, Li L, Goldstein D, Burge M, Cooray P, Nagrial A, Tebbutt NC, Thomson B, Nikfarjam M, Harris M, Haydon A, Lawrence B, Tai DWM, Simons K, Lennon AM, Wolfgang CL, Tomasetti C, Papadopoulos N, Kinzler KW, Vogelstein B, Gibbs P. Circulating tumor DNA as a potential marker of adjuvant chemotherapy benefit following surgery for localized pancreatic cancer. Ann Oncol. 2019;30:1472-1478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 118]  [Article Influence: 23.6]  [Reference Citation Analysis (0)]
127.  Pietrasz D, Pécuchet N, Garlan F, Didelot A, Dubreuil O, Doat S, Imbert-Bismut F, Karoui M, Vaillant JC, Taly V, Laurent-Puig P, Bachet JB. Plasma Circulating Tumor DNA in Pancreatic Cancer Patients Is a Prognostic Marker. Clin Cancer Res. 2017;23:116-123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 170]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
128.  Okada T, Mizukami Y, Ono Y, Sato H, Hayashi A, Kawabata H, Koizumi K, Masuda S, Teshima S, Takahashi K, Katanuma A, Omori Y, Iwano H, Yamada M, Yokochi T, Asahara S, Kawakubo K, Kuwatani M, Sakamoto N, Enomoto K, Goto T, Sasajima J, Fujiya M, Ueda J, Matsumoto S, Taniue K, Sugitani A, Karasaki H, Okumura T. Digital PCR-based plasma cell-free DNA mutation analysis for early-stage pancreatic tumor diagnosis and surveillance. J Gastroenterol. 2020;55:1183-1193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
129.  Park Y, Jun HR, Choi HW, Hwang DW, Lee JH, Song KB, Lee W, Kwon J, Ha SH, Jun E, Kim SC. Circulating tumour cells as an indicator of early and systemic recurrence after surgical resection in pancreatic ductal adenocarcinoma. Sci Rep. 2021;11:1644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
130.  Allenson K, Castillo J, San Lucas FA, Scelo G, Kim DU, Bernard V, Davis G, Kumar T, Katz M, Overman MJ, Foretova L, Fabianova E, Holcatova I, Janout V, Meric-Bernstam F, Gascoyne P, Wistuba I, Varadhachary G, Brennan P, Hanash S, Li D, Maitra A, Alvarez H. High prevalence of mutant KRAS in circulating exosome-derived DNA from early-stage pancreatic cancer patients. Ann Oncol. 2017;28:741-747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 344]  [Cited by in F6Publishing: 332]  [Article Influence: 47.4]  [Reference Citation Analysis (0)]
131.  Takahasi K, Iinuma H, Wada K, Minezaki S, Kawamura S, Kainuma M, Ikeda Y, Shibuya M, Miura F, Sano K. Usefulness of exosome-encapsulated microRNA-451a as a minimally invasive biomarker for prediction of recurrence and prognosis in pancreatic ductal adenocarcinoma. J Hepatobiliary Pancreat Sci. 2018;25:155-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 97]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
132.  Kawamura S, Iinuma H, Wada K, Takahashi K, Minezaki S, Kainuma M, Shibuya M, Miura F, Sano K. Exosome-encapsulated microRNA-4525, microRNA-451a and microRNA-21 in portal vein blood is a high-sensitive liquid biomarker for the selection of high-risk pancreatic ductal adenocarcinoma patients. J Hepatobiliary Pancreat Sci. 2019;26:63-72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
133.  Del Re M, Vivaldi C, Rofi E, Vasile E, Miccoli M, Caparello C, d'Arienzo PD, Fornaro L, Falcone A, Danesi R. Early changes in plasma DNA levels of mutant KRAS as a sensitive marker of response to chemotherapy in pancreatic cancer. Sci Rep. 2017;7:7931.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 54]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
134.  Kruger S, Heinemann V, Ross C, Diehl F, Nagel D, Ormanns S, Liebmann S, Prinz-Bravin I, Westphalen CB, Haas M, Jung A, Kirchner T, von Bergwelt-Baildon M, Boeck S, Holdenrieder S. Repeated mutKRAS ctDNA measurements represent a novel and promising tool for early response prediction and therapy monitoring in advanced pancreatic cancer. Ann Oncol. 2018;29:2348-2355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 80]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
135.  Wei T, Zhang X, Zhang Q, Yang J, Chen Q, Wang J, Li X, Chen J, Ma T, Li G, Gao S, Lou J, Que R, Wang Y, Dang X, Zheng L, Liang T, Bai X. Vimentin-positive circulating tumor cells as a biomarker for diagnosis and treatment monitoring in patients with pancreatic cancer. Cancer Lett. 2019;452:237-243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 72]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
136.  Tian X, Shivapurkar N, Wu Z, Hwang JJ, Pishvaian MJ, Weiner LM, Ley L, Zhou D, Zhi X, Wellstein A, Marshall JL, He AR. Circulating microRNA profile predicts disease progression in patients receiving second-line treatment of lapatinib and capecitabine for metastatic pancreatic cancer. Oncol Lett. 2016;11:1645-1650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
137.  An M, Lohse I, Tan Z, Zhu J, Wu J, Kurapati H, Morgan MA, Lawrence TS, Cuneo KC, Lubman DM. Quantitative Proteomic Analysis of Serum Exosomes from Patients with Locally Advanced Pancreatic Cancer Undergoing Chemoradiotherapy. J Proteome Res. 2017;16:1763-1772.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 79]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
138.  Yin L, Pu N, Thompson E, Miao Y, Wolfgang C, Yu J. Improved Assessment of Response Status in Patients with Pancreatic Cancer Treated with Neoadjuvant Therapy using Somatic Mutations and Liquid Biopsy Analysis. Clin Cancer Res. 2021;27:740-748.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
139.  Gemenetzis G, Groot VP, Yu J, Ding D, Teinor JA, Javed AA, Wood LD, Burkhart RA, Cameron JL, Makary MA, Weiss MJ, He J, Wolfgang CL. Circulating Tumor Cells Dynamics in Pancreatic Adenocarcinoma Correlate With Disease Status: Results of the Prospective CLUSTER Study. Ann Surg. 2018;268:408-420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 111]  [Article Influence: 18.5]  [Reference Citation Analysis (1)]
140.  Poruk KE, Valero V 3rd, Saunders T, Blackford AL, Griffin JF, Poling J, Hruban RH, Anders RA, Herman J, Zheng L, Rasheed ZA, Laheru DA, Ahuja N, Weiss MJ, Cameron JL, Goggins M, Iacobuzio-Donahue CA, Wood LD, Wolfgang CL. Circulating Tumor Cell Phenotype Predicts Recurrence and Survival in Pancreatic Adenocarcinoma. Ann Surg. 2016;264:1073-1081.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 109]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
141.  Neumann MHD, Bender S, Krahn T, Schlange T. ctDNA and CTCs in Liquid Biopsy - Current Status and Where We Need to Progress. Comput Struct Biotechnol J. 2018;16:190-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 136]  [Article Influence: 22.7]  [Reference Citation Analysis (0)]
142.  Ding Y, Li W, Wang K, Xu C, Hao M, Ding L. Perspectives of the Application of Liquid Biopsy in Colorectal Cancer. Biomed Res Int. 2020;2020:6843180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
143.  Werner S, Keller L, Pantel K. Epithelial keratins: Biology and implications as diagnostic markers for liquid biopsies. Mol Aspects Med. 2020;72:100817.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 41]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
144.  Heidrich I, Ačkar L, Mossahebi Mohammadi P, Pantel K. Liquid biopsies: Potential and challenges. Int J Cancer. 2021;148:528-545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 127]  [Article Influence: 31.8]  [Reference Citation Analysis (0)]
145.  Fattore L, Ruggiero CF, Liguoro D, Castaldo V, Catizone A, Ciliberto G, Mancini R. The Promise of Liquid Biopsy to Predict Response to Immunotherapy in Metastatic Melanoma. Front Oncol. 2021;11:645069.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]