Next Article in Journal
Synthesis of Hantzsch Adducts as Cholinesterases and Calcium Flux inhibitors, Antioxidants and Neuroprotectives
Next Article in Special Issue
ANK2 Hypermethylation in Canine Mammary Tumors and Human Breast Cancer
Previous Article in Journal
Nitroxides Mitigate Neutrophil-Mediated Damage to the Myocardium after Experimental Myocardial Infarction in Rats
Previous Article in Special Issue
Unique Interplay between Molecular miR-181b/d Biomarkers and Health Related Quality of Life Score in the Predictive Glioma Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Status of Circulating Tumor DNA Liquid Biopsy in Pancreatic Cancer

1
Department of Surgery, Rush University Medical Center, Chicago, IL 60612, USA
2
Division of Hematology/Oncology and Cell Therapy, Rush University Medical Center, Chicago, IL 60612, USA
3
Rush Precision Oncology Program, Rush University Medical Center, Chicago, IL 60612, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(20), 7651; https://doi.org/10.3390/ijms21207651
Submission received: 16 September 2020 / Revised: 12 October 2020 / Accepted: 14 October 2020 / Published: 16 October 2020
(This article belongs to the Special Issue Circulating Cell-Free Nucleic Acids and Cancers)

Abstract

:
Pancreatic cancer is a challenging disease with a low 5-year survival rate. There are areas for improvement in the tools used for screening, diagnosis, prognosis, treatment selection, and assessing treatment response. Liquid biopsy, particularly cell free DNA liquid biopsy, has shown promise as an adjunct to our standard care for pancreatic cancer patients, but has not yet been universally adopted into regular use by clinicians. In this publication, we aim to review cfDNA liquid biopsy in pancreatic cancer with an emphasis on current techniques, clinical utility, and areas of active investigation. We feel that researchers and clinicians alike should be familiar with this exciting modality as it gains increasing importance in the care of cancer patients.

1. Pancreatic Cancer: The Need for a Novel Diagnostic

Pancreatic ductal adenocarcinoma (PDAC) has an incidence of 13.1 cases per 100,000 persons in the United States, where it is currently the third leading cause of cancer mortality, and is expected to rise [1]. Five-year overall survival (OS) across all stages is a dismal 10% [1,2,3,4]. Early stage disease can be treated with curative intent, however, 5-year OS even in this case is 20–40% [5,6,7]. Poor survival in PDAC is attributed to advanced stage at presentation. Thus, in addition to the development of more effective treatment strategies for metastatic disease, the key to improve survival is early detection and implementation of curative-intent therapy.
Early detection of PDAC is limited by issues related to the tumor itself and the technology available for diagnosis. PDAC is often clinically silent in the early phases; only 10–20% of patients with PDAC are candidates for curative-intent therapy at time of diagnosis [8,9]. Presently, the United States Preventive Service Task Force (USPSTF) recommends against screening of the general population for PDAC owing to lack of an appropriate diagnostic assay. Available tests are neither sensitive nor specific for definitive diagnosis, and it is perceived that the harm of population level screening would outweigh the benefits of early detection [10,11].
The diagnosis of PDAC is currently confirmed via endoscopic ultrasound (EUS) with fine-needle aspiration for cytologic analysis [12,13]. Management is multimodal and depends chiefly on staging with EUS and radiographic cross-sectional imaging, often with computed tomography (CT) [8]. The decision to pursue or forgo surgical resection and curative intent therapy is a critical branch point in all treatment algorithms. Multidisciplinary tumor boards stratify patients into surgically resectable, borderline resectable, or unresectable subgroups based on the presence of distal disease and/or local tumor advancement [14]. Following treatment with either neoadjuvant chemotherapy, upfront surgery, or palliative intent systemic therapy, patients undergo serial evaluation with imaging and CA 19-9 analysis for surveillance in a curative setting or to ascertain response to therapies in a palliative setting [8,15,16,17].
Liquid biopsy is an emerging technology that permits the noninvasive sampling of tumor material in circulation [18]. It is under investigation for a number of gastrointestinal malignancies and is believed to offer immediate and theoretical advantages over current standards of care in several aspects of oncologic assessment (Figure 1) [19,20]. Our aim is to outline the present status of liquid biopsy as it relates to the management of PDAC with a focus on the use of cell free DNA (cfDNA) and circulating tumor DNA (ctDNA).

2. cfDNA and ctDNA in PDAC

The term “cfDNA” refers to free DNA, usually within the circulatory system, derived from both benign and malignant cells. “ctDNA” is more specific and refers to the tumor-derived portion of the cfDNA [18]. The principle behind peripherally detected cfDNA is that, as malignant and benign cells undergo apoptosis or necrosis, genetic material is released into circulation as either free nucleic acids or material that has remained encapsulated within the bounds of a partial or complete cell membrane [21,22,23,24,25]. The history of cfDNA is fairly recent; it was first isolated from human plasma in 1948 [26]. By 1965, a link between cfDNA and malignant processes had been established [27].
Liquid biopsy, broadly, uses one or more circulating biomarkers to assess a disease process. In the case of PDAC, much attention has been given to cfDNA, ctDNA, tumor-derived exosomes, and circulating tumor cells (CTCs) [28,29]. The ctDNA can provide clinicians information regarding PDAC as it harbors mutations or epigenetic characteristics specific to the patient’s cancerous process. The advancement of cfDNA and liquid biopsy-based cancer research has been largely dependent on parallel advances in oncogenetics and genomics, as these fields have characterized the key mutations that could be detected in peripheral blood samples.
Mutations present in the primary tumor, metastatic lesions, or both are detectable and sometimes distinguishable from one another. Mutations not detected on tissue biopsy because of intratumoral heterogeneity and sampling error may be detectable on a liquid biopsy. Low concordance between primary tumor and ctDNA collected from the periphery has been demonstrated [24]. Assessment of liquid biopsy derived genetic material has the potential to contribute to the diagnosis, staging, and surveillance of PDAC or potentially guide precision targeted therapy based on the specific mutation identified.
The identification and characterization of KRAS mutations as they relate to PDAC began in the 1980s. KRAS is often a founding mutation in pancreatic cancer, present in over 90% of primary tumors, and thus has become a primary target for analysis in ctDNA-based assays [30]. Additional, common mutations identified in the ctDNA of patients with PDAC include CDKN2A, SMAD4, and TP53. Further, actionable alterations in the RTK-RAS-RAF pathway including BRAF mutations, ERBB2 mutations, and FGFR1 amplifications are found in a smaller percentage of PDAC [31]. Tumors without KRAS mutations may harbor other mutations that can be detected with modern amplification and sequencing techniques [24].

A Brief Mention of Exosomes

Exosomes are extracellular vesicles, typically between 50 and 150 nanometers in size, derived from cells (including cancer cells) that contain proteins and genetic material that may be isolated and analyzed. Theoretical advantages of exosome analysis in liquid biopsy are that exosomes have a longer circulating half-life than ctDNA and cancer cells are constantly producing exosomes so peripheral circulation capture may not be dependent on events such as cell necrosis, apoptosis, or invasion. These characteristics may allow for earlier detection [32,33,34].
Exosomes can be detected in many different fluids including effusions, ascites, blood, and saliva. Within the discipline of PDAC care, exosomes are being actively explored, but are not currently in widespread clinical use. Protein and genetic analysis may prove useful in diagnostics, screening, and targeted therapy [35,36,37].

3. Liquid Biopsy Techniques in PDAC

cfDNA-based liquid biopsy is a multistep process involving sample collection, processing, and polymerase chain reaction (PCR)-based nucleic acid analysis. The overarching processes are well described. While laboratory protocols are generally standardized, some variation exists in isolation/purification, PCR, and sequencing (Figure 2) [29].
One important area of this process is the isolation and purification of the cfDNA. Two techniques have predominated this crucial part of cfDNA analysis, spin column or magnetic bead. At this juncture, the spin column technique is considered the gold standard, with slightly higher yields. DNA isolation may be one of the limiting factors in PDAC liquid biopsy as early disease frequently has exceptionally low levels of peripheral ctDNA, making a meaningful yield challenging [38].
Amplification/quantification follows the isolation/purification steps. In the case of PDAC, KRAS mutations are a common target, as over 90% of PDAC contains at least one KRAS mutation in tissue samples [39]. Of KRAS-mutated cancers, 90% will have one of four common polymorphisms: G12D, G12V, or G12R (40, 36, and 12 percent, respectively, for tissue samples) [40,41]. Various PCR protocols have been utilized including quantitative and droplet/digital PCR for amplification [42].
Sequencing can occur following amplification. Recently, high-throughput next generation sequencing (NGS)/massively parallel sequencing (MPS) has been utilized in ctDNA-based liquid biopsy [40,43,44,45]. Both entire ctDNA sequencing and targeted gene sequencing have been attempted [45,46]. This type of modality may be particularly useful in screening for actionable gene mutations and the advancement of precision oncology [47]. Success has been mostly demonstrated in patients with high tumor burden, and thus a higher percentage of ctDNA in circulation. Studies utilizing NGS have covered over 20,000 genes (and sometimes target specific genes); coverage (depth) has been variable, but 234× to 2227× has been reported in the literature [46,48].
CTCs and exosomes can be analyzed in a similar fashion to cfDNA. Flow cytometry, electron microscopy, or Western blot analysis can all be utilized in the identification of exosomes. However, unlike cfDNA, exosomes require ultracentrifugation for separation from interfering blood components [49]. Next, membranes are lysed and DNA/RNA can be isolated. In addition to analysis of nucleic acids, exosomes and CTCs can undergo quantification, protein analysis, and transcriptomics, providing additional information to researchers and clinicians [50].

4. Current Clinical Utility of cfDNA

4.1. ctDNA as a Screening Test

Screening for PDAC has the potential to improve outcomes if early stage disease can be detected; localized cancer has a five-year OS of 20% compared with metastatic PDAC’s 3% [2]. No PDAC screening tests are currently available for the average risk person and active investigation into the utility of liquid biopsy, specifically with ctDNA, is underway.
A meta-analysis including seven studies of ctDNA using various isolation protocols, gene loci, and epigenetic markers demonstrated a pooled sensitivity of only 0.64 [51]. The highest recorded sensitivity in an individual study was greater than 0.95 and the lowest was 0.27 [52,53,54,55,56]. The most widely accepted hypothesis underlying the low observed sensitivity is that, in early PDAC, there is an inadequate quantity of tumor necrosis necessary to release a detectable amount of peripheral ctDNA [30]. In the early stages of PDAC, there may only be one molecule of ctDNA per every 5 mL of plasma [51].
Another issue is the 20% rate of false positives derived from circulating KRAS mutations in patients with chronic pancreatitis [33]. On the other hand, used in combination with CA19-9 in a study of 47 patients, ctDNA was able to differentiate PDAC from chronic pancreatitis with a sensitivity of 98% and a specificity of 77% [57]. Multitarget screening tests may be more sensitive and have shown promise [58,59,60]. Advancement in techniques for increasing ctDNA yields will be necessary for the development of a ctDNA-based PDAC screening test. New research in epigenetics, specifically DNA methylation, has shown promise in improving sensitivity, however, larger and more comprehensive prospective studies are warranted [54,56,61,62].

4.2. ctDNA as a Diagnostic and Prognostic Test

Currently, diagnostic tests for PDAC include the combination of cross-sectional imaging and image-guided biopsy, usually an EUS guided fine-needle aspiration. These biopsies are invasive; unpleasant for the patient; and carry the risks of anesthesia, perforation, and infection. One of the endeavors of liquid biopsy would be to minimize the need for such invasive tests.
Compared with EUS, ctDNA offers increased convenience and lower risk. A recent meta-analysis demonstrated a pooled specificity for ctDNA in detection of PDAC of 0.92. This utility is limited in early disease, as mentioned previously. The current standard biomarker, CA19-9, has a specificity for PDAC of over 90% at certain cutoff values, thus, with current technology, ctDNA offers only a marginal if any improvement (regarding assessing whether or not PDAC is present) [52,63]. The use of ctDNA liquid biopsy in conjunction with traditional biomarkers has shown some promise in improving sensitivity and specificity in early PDAC [58]. However, if current trends continue and the diagnostic accuracy of ctDNA-based liquid biopsy continues to improve, one could envision a time when the need for invasive tissue biopsy of the primary lesion is obviated.
Potential advantages of ctDNA-based liquid biopsy are seen in its ability to prognosticate, as well as to diagnose actionable mutations in a minimally invasive fashion. Regarding its prognostic capabilities, current research has demonstrated that greater than 0.6% ctDNA was predictive of an OS of 6.3 months, while less than 0.6% had an OS of 11.7 months [24]. Peripherally detected KRAS mutations are a poor prognostic indicator at all stages of disease [64]. Mutated KRAS in the peripheral circulation has demonstrated worse OS (3 vs. 11 months in some studies) [65,66,67,68,69].
Regarding the diagnosis of actionable mutations for targeted therapy, ctDNA-based liquid biopsy has shown great promise. This is particularly important in the setting of metastatic disease where targeted therapies are being implemented for patients who have progressed while receiving standard of care regimens. It is thought that ctDNA has a higher concordance with metastatic lesions than primary tumor tissue [24,70,71]. Metastatic lesions may contain targetable mutations that are not present within the biopsy of the primary site. Liquid biopsy may diagnose these mutations, broadening treatment options and sparing additional biopsies for the sickest of PDAC patients. Within this context, ctDNA is being actively explored as a means to guide systemic treatment [72,73]. Common targetable mutations in metastatic PDAC include specific KRAS mutations, BRAF, and ERBB2, among others [73,74]. At present, this theoretical advantage has not yet translated into a quantifiable clinical benefit.

4.3. ctDNA in Assessing Resectability of Primary Tumor

The determination of resectability is key to the multidisciplinary management of PDAC. The principal determinants of resectability are (1) the degree of local mesenteric vascular involvement and (2) the presence of distant metastases [8]. EUS and pancreas protocol CT have an excellent diagnostic yield in evaluating the anatomic relationships to mesenteric vessels that dictate resectability, thus the role of ctDNA in this respect may be limited [75]. Higher levels of peripheral KRAS mutation have been associated with the presence of direct mesenteric vascular involvement, but with a low degree of sensitivity and specificity [65,66]. Portal venous sampling is feasible and has a higher ctDNA yield than peripheral blood, however, the relationship of portal ctDNA levels to local vascular invasion is unknown. Further, portal venous sampling is invasive and negates the theoretical advantage of a noninvasive peripheral blood liquid biopsy.
On the other hand, room for improvement exists in the detection of systemic disease that would preclude attempts at resection. The 80% incidence of distant metastases within 5 years of an R0 resection implies that no contemporary test adequately detects systemic disease in the preoperative period [76,77]. The majority of metastases occur in the liver or peritoneal surface, where sub-centimeter malignant implants are difficult to detect radiographically [22]. Current staging protocols involve chest and pelvic CT, magnetic resonance imaging (MRI) or positron emission tomography (PET) of the abdomen to evaluate indeterminant liver lesions, and in some cases, staging laparoscopy. Elevated traditional biomarkers such as CA19-9 are predictive of occult metastases, albeit with a false positive rate up to 47% [78]. A study of 1001 patients since 2000 demonstrated that even laparoscopy, which requires operating room time and general anesthesia, missed between 10 and 30% of occult metastases identified during open surgery, without mention of early recurrences in resected patients [79]. Discordance between mutations detected in ctDNA versus tissue biopsy may indicate the presence of radiographically occult metastases. The fact that ctDNA is more concordant with mutations identified in tissue biopsy from metastatic lesions than the primary tumor supports this [24]. However, intratumoral heterogeneity and sampling error in the primary tissue biopsy partially account for this observation [80].
ctDNA-based liquid biopsy has the potential to assist in the detection of occult metastatic disease. This could spare patients the morbidity of an operation from which they would reap no benefit. A study of 112 patients demonstrated that those with resectable disease had both a lower percent ctDNA and fewer genomic alterations in ctDNA than patients with unresectable disease [24,81].
As KRAS is mutated in over 90% of PDAC and is reliably detectable in inexpensive assays, ctDNA-based liquid biopsy designed to detect mutations at this locus for staging purposes can be a feasible strategy if confirmed in a prospective large study [82]. A study of 151 patients demonstrated peripherally detected KRAS mutations in 58.9% of patients with radiographic distant organ metastasis compared with 18.2% of patients without them. In the same study, only 9 out of 108 (8.3%) patients with successfully resected disease had peripherally detected KRAS mutations. Only 4.6 percent of patients with a negative preoperative ctDNA-based liquid biopsy insuffered distant recurrences within 6 months of surgery, compared with 35% in a separate study with radiographically defined resectable disease, but no liquid biopsy testing [83,84]. Thus, radiographic staging and then further stratification with ctDNA may optimize the selection of patients who would benefit from surgical resection. A study of 23 patients with detectable ctDNA prior to surgical resection demonstrated that 12 converted to ctDNA-negative in the postoperative period. Of these 12, median recurrence free survival was inferior to those with undetectable preoperative ctDNA at 12.2 months compared with over 38 months in the preoperatively negative ctDNA group [85]. Early data have demonstrated that patients who have undergone neoadjuvant chemotherapy and are ctDNA negative have an 80% chance of an R0, node negative resection compared with those with who are ctDNA positive, having only a 38% chance of an R0, node negative operation [86]. Taken together, these data indicate that patients with radiographically resectable disease may fall, broadly, within two subsets: those who are ctDNA positive with worse outcomes and those who are ctDNA negative with better outcomes [87,88,89].
Preoperative ctDNA positivity clearly indicates a high risk of postoperative recurrence; however, further prospective research is required to determine whether it is a hard indicator of surgical futility. Ultimately, based on the combined results of preoperative ctDNA and traditional imaging modalities, patients could either be spared the morbidity of a laparotomy in cases where it is futile, or referred for neoadjuvant therapy and restaged.

4.4. Surveillance

Recurrence after curative-intent therapy is high in PDAC. One study of patients with early stage, resectable disease demonstrated 35% recurrence at 1-year follow up, while 1-year recurrence rates over 80% have been observed in patients with borderline resectable disease [84,90]. Early detection of recurrences is key to initiating appropriate systemic therapy. Surveillance for recurrence or progression of PDAC involves traditional tumor markers such as CA19-9 and cross-sectional imaging. ctDNA may be superior to current techniques in the sense that it offers earlier detection and the opportunity to guide targeted therapy [74]. The short half-life of ctDNA in circulation (measured in hours) is ideal for assessing response to therapy in shorter intervals [91].
In 10 patients who recurred after curative-intent therapy, ctDNA-based liquid biopsy was able to detect disease progression at 3.1 months, compared with 9.6 months with standard CT-based surveillance [48]. Postoperatively detectable ctDNA had a 100% positive predictive value for disease recurrence with a sensitivity of 57%. While 13 of 13 patients with postoperatively detectable ctDNA relapsed, only 10 of 22 patients with undetectable postoperative ctDNA had recurrences at 38 months post-resection [92]. CA19-9 has traditionally been used as a surveillance tool, but has its limitations, namely low sensitivity and the fact that 5–10% of the population are incapable of producing CA19-9, rendering it useless in these patients [93,94]. ctDNA can be used for monitoring throughout treatments including surgery and chemotherapy [95,96,97,98,99].
Future application of ctDNA in surveillance is an exciting area of investigation. Further incorporating ctDNA guided surveillance in large prospective studies may aid in the development of risk-adjusted treatment strategies for adjuvant chemotherapy. For instance, a less aggressive chemotherapy regimen may be appropriate for patients who are ctDNA negative, sparing them from unpleasant toxicities, whereas patients with persistent ctDNA on completion on adjuvant therapy may need close surveillance so that recurrence is detected early to improve the clinical outcomes.

4.5. Clinical Utility Of Exosomes

Exosomes remain an area of active exploration. Both genetic and protein components of exosomes have been assessed for utility in PDAC. In particular, glypican 1 expression has been proposed as an adjunct for PDAC diagnosis [35,36,100]. PDAC-associated KRAS mutations have been reliably identified in exosome derived RNA/DNA [33,36,101]. However, similar to cfDNA, early detection remains challenging and limits utility as a screening tool [33]. There remains a paucity of research on the utility of exosomal analysis for tracking response to treatment or as a prognostic tool in PDAC.

5. Ongoing Clinical Trials and the Future of Cfdna in Pdac

While not the standard of care at present, cfDNA-based liquid biopsy in its current form can contribute to management of PDAC and a wide range of new indications for its use are under development. Current trials are exploring screening, prognostics, precision oncology, and targeted therapy. Table 1 lists current trials registered with clinicaltrials.gov. cfDNA-based liquid biopsy is an exciting field that has applications for multiple cancers, the scope of which is not covered in this review, yet advances in other fields will likely come with parallel progress in liquid biopsy for PDAC.

6. Conclusions

PDAC remains a deadly and challenging cancer for patients and clinicians, respectively. There are opportunities for improvement in screening, treatment guidance, and surveillance. The use of ctDNA as a liquid biopsy is an area of active and fruitful investigation.
One of the most valuable areas in which ctDNA can be utilized is as a guide toward or away from surgical resection based on detection of ctDNA, typically by the presence of mutated KRAS. Microscopic metastases have been a persistent problem in the care of PDAC, and ctDNA may be the tool that helps identify patients with this condition prior to receiving unwarranted therapy. Thus, for radiographically resectable disease, the utility of ctDNA lies in the identification of patients appropriate for neoadjuvant therapy, and in sparing certain patients the morbidity of major abdominal surgery in cases where it would be futile. Multiple small studies have demonstrated the utility of ctDNA as a tool for monitoring response to different treatments. As novel, targeted therapies continue to emerge, the role of genetic assessment for these targets will play an ever-larger role in cancer care. ctDNA analysis with NGS seems to be a promising field and should be further explored in the setting of PDAC.
ctDNA-based liquid biopsy has potential to be implemented in multiple phases of care in patients with pancreatic cancer if confirmed in large prospective studies. We anticipate adoption into the clinician’s tool kit over the next several years once current clinical trials come to conclusion.

Author Contributions

Conceptualization, M.W.G., R.A.J., S.G.P., and A.M.; writing—original draft preparation, M.W.G. and R.A.J.; writing—review and editing, S.G.P., A.M., and T.M.K.; supervision, S.G.P. and A.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors would like to acknowledge the Rush University Medical Center’s Department of Surgery and Precision Oncology Program for their support and guidance.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

PDACPancreatic Ductal Adenocarcinoma
USPSTFUnited States Preventative Service Task Force
OSOverall Survival
EUSEndoscopic Ultrasound
cfDNACell Free Deoxyribonucleic Acid
ctDNACell Free Tumor Deoxyribonucleic Acid
CTCsCirculating Tumor Cells
PCRPolymerase Chain Reaction
NGSNext Generation Sequencing

References

  1. Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting Cancer Incidence and Deaths to 2030: The Unexpected Burden of Thyroid, Liver, and Pancreas Cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef] [Green Version]
  2. SEER Cancer Statistics Review, 1975–2017. Available online: https://seer.cancer.gov/csr/1975_2017/index.html (accessed on 13 July 2020).
  3. Ilic, M.; Ilic, I. Epidemiology of pancreatic cancer. World J. Gastroenterol. 2016, 22, 9694–9705. [Google Scholar] [CrossRef]
  4. Chang, J.S.; Chen, L.; Shan, Y.; Chu, P.; Tsai, C.; Tsai, H. The incidence and survival of pancreatic cancer by histology, including rare subtypes: A nation-wide cancer registry-based study from Taiwan. Cancer Med. 2018, 7, 5775–5788. [Google Scholar] [CrossRef] [Green Version]
  5. Huang, L.; Jansen, L.; Balavarca, Y.; Babaei, M.; van der Geest, L.; Lemmens, V.; Van Eycken, L.; De Schutter, H.; Johannesen, T.B.; Primic-Žakelj, M.; et al. Stratified survival of resected and overall pancreatic cancer patients in Europe and the USA in the early twenty-first century: A large, international population-based study. BMC Med. 2018, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Shaib, Y.; Davila, J.; Naumann, C.; El-Serag, H. The Impact of Curative Intent Surgery on the Survival of Pancreatic Cancer Patients: A U.S. Population-Based Study. Off. J. Am. Coll. Gastroenterol. ACG 2007, 102, 1377–1382. [Google Scholar] [CrossRef]
  7. Mokdad, A.A.; Minter, R.M.; Zhu, H.; Augustine, M.M.; Porembka, M.R.; Wang, S.C.; Yopp, A.C.; Mansour, J.C.; Choti, M.A.; Polanco, P.M. Neoadjuvant Therapy Followed by Resection Versus Upfront Resection for Resectable Pancreatic Cancer: A Propensity Score Matched Analysis. J. Clin. Oncol. 2017, 35, 515–522. [Google Scholar] [CrossRef]
  8. Vincent, A.; Herman, J.; Schulick, R.; Hruban, R.H.; Goggins, M. Pancreatic cancer. Lancet 2011, 378, 607–620. [Google Scholar] [CrossRef]
  9. Gobbi, P.G.; Bergonzi, M.; Comelli, M.; Villano, L.; Pozzoli, D.; Vanoli, A.; Dionigi, P. The prognostic role of time to diagnosis and presenting symptoms in patients with pancreatic cancer. Cancer Epidemiol. 2013, 37, 186–190. [Google Scholar] [CrossRef]
  10. Force, U.P.S.T.; Owens, D.K.; Davidson, K.W.; Krist, A.H.; Barry, M.J.; Cabana, M.; Caughey, A.B.; Curry, S.J.; Doubeni, C.A.; Epling, J.W.; et al. Screening for Pancreatic Cancer: US Preventive Services Task Force Reaffirmation Recommendation Statement. JAMA 2019, 322, 438–444. [Google Scholar] [CrossRef] [Green Version]
  11. McAllister, F.; Montiel, M.F.; Uberoi, G.S.; Uberoi, A.S.; Maitra, A.; Bhutani, M.S. Current Status and Future Directions for Screening Patients at High Risk for Pancreatic Cancer. Gastroenterol. Hepatol. (N. Y.) 2017, 13, 268–275. [Google Scholar]
  12. Kitano, M.; Yoshida, T.; Itonaga, M.; Tamura, T.; Hatamaru, K.; Yamashita, Y. Impact of endoscopic ultrasonography on diagnosis of pancreatic cancer. J. Gastroenterol. 2019, 54, 19–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Cheng, B.; Zhang, Y.; Chen, Q.; Sun, B.; Deng, Z.; Shan, H.; Dou, L.; Wang, J.; Li, Y.; Yang, X.; et al. Analysis of Fine-Needle Biopsy vs Fine-Needle Aspiration in Diagnosis of Pancreatic and Abdominal Masses: A Prospective, Multicenter, Randomized Controlled Trial. Clin. Gastroenterol. Hepatol. 2018, 16, 1314–1321. [Google Scholar] [CrossRef] [PubMed]
  14. Wittel, U.A.; Lubgan, D.; Ghadimi, M.; Belyaev, O.; Uhl, W.; Bechstein, W.O.; Grützmann, R.; Hohenberger, W.M.; Schmid, A.; Jacobasch, L.; et al. Consensus in determining the resectability of locally progressed pancreatic ductal adenocarcinoma—Results of the Conko-007 multicenter trial. BMC Cancer 2019, 19, 979. [Google Scholar] [CrossRef] [PubMed]
  15. Neoptolemos, J.P.; Stocken, D.D.; Friess, H.; Bassi, C.; Dunn, J.A.; Hickey, H.; Beger, H.; Fernandez-Cruz, L.; Dervenis, C.; Lacaine, F.; et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N. Engl. J. Med. 2004, 350, 1200–1210. [Google Scholar] [CrossRef] [Green Version]
  16. Conroy, T.; Desseigne, F.; Ychou, M.; Bouché, O.; Guimbaud, R.; Bécouarn, Y.; Adenis, A.; Raoul, J.-L.; Gourgou-Bourgade, S.; de la Fouchardière, C.; et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N. Engl. J. Med. 2011, 364, 1817–1825. [Google Scholar] [CrossRef] [Green Version]
  17. Ferrone, C.R.; Marchegiani, G.; Hong, T.S.; Ryan, D.P.; Deshpande, V.; McDonnell, E.I.; Sabbatino, F.; Santos, D.D.; Allen, J.N.; Blaszkowsky, L.S.; et al. Radiological and surgical implications of neoadjuvant treatment with FOLFIRINOX for locally advanced and borderline resectable pancreatic cancer. Ann. Surg. 2015, 261, 12–17. [Google Scholar] [CrossRef] [Green Version]
  18. Marrugo-Ramírez, J.; Mir, M.; Samitier, J. Blood-Based Cancer Biomarkers in Liquid Biopsy: A Promising Non-Invasive Alternative to Tissue Biopsy. Int. J. Mol. Sci. 2018, 19, 2877. [Google Scholar] [CrossRef] [Green Version]
  19. Crowley, E.; Di Nicolantonio, F.; Loupakis, F.; Bardelli, A. Liquid biopsy: Monitoring cancer-genetics in the blood. Nat. Rev. Clin. Oncol. 2013, 10, 472–484. [Google Scholar] [CrossRef]
  20. Vymetalkova, V.; Cervena, K.; Bartu, L.; Vodicka, P. Circulating Cell-Free DNA and Colorectal Cancer: A Systematic Review. Int. J. Mol. Sci. 2018, 19, 3356. [Google Scholar] [CrossRef] [Green Version]
  21. Yadav, D.K.; Bai, X.; Yadav, R.K.; Singh, A.; Li, G.; Ma, T.; Chen, W.; Liang, T. Liquid biopsy in pancreatic cancer: The beginning of a new era. Oncotarget 2018, 9, 26900–26933. [Google Scholar] [CrossRef] [Green Version]
  22. Jacobson, R.A.; Munding, E.; Hayden, D.M.; Levy, M.; Kuzel, T.M.; Pappas, S.G.; Masood, A. Evolving Clinical Utility of Liquid Biopsy in Gastrointestinal Cancers. Cancers 2019, 11, 1164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. De Wit, S.; van Dalum, G.; Terstappen, L.W.M.M. Detection of Circulating Tumor Cells. Scientifica (Cairo) 2014, 2014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Patel, H.; Okamura, R.; Fanta, P.; Patel, C.; Lanman, R.B.; Raymond, V.M.; Kato, S.; Kurzrock, R. Clinical correlates of blood-derived circulating tumor DNA in pancreatic cancer. J. Hematol. Oncol. 2019, 12, 130. [Google Scholar] [CrossRef] [Green Version]
  25. Alix-Panabières, C.; Pantel, K. Circulating Tumor Cells: Liquid Biopsy of Cancer. Clin. Chem. 2013, 59, 110–118. [Google Scholar] [CrossRef] [PubMed]
  26. Mandel, P.; Metais, P. Les acides nucleiques du plasma sanguin chez l’ homme. C. R. Seances Soc. Biol. Fil. 1948, 142, 241–243. [Google Scholar]
  27. Bendich, A.; Wilczok, T.; Borenfreund, E. Circulating DNA as a Possible Factor in Oncogenesis. Science 1965, 148, 374–376. [Google Scholar] [CrossRef] [PubMed]
  28. Samandari, M.; Julia, M.G.; Rice, A.; Chronopoulos, A.; Armando, E. Liquid biopsies for management of pancreatic cancer. Transl. Res. 2018, 201, 98–127. [Google Scholar] [CrossRef]
  29. Merker, J.D.; Oxnard, G.R.; Compton, C.; Diehn, M.; Hurley, P.; Lazar, A.J.; Lindeman, N.; Lockwood, C.M.; Rai, A.J.; Schilsky, R.L.; et al. Circulating Tumor DNA Analysis in Patients With Cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. JCO 2018, 36, 1631–1641. [Google Scholar] [CrossRef]
  30. Kinugasa, H.; Nouso, K.; Miyahara, K.; Morimoto, Y.; Dohi, C.; Tsutsumi, K.; Kato, H.; Matsubara, T.; Okada, H.; Yamamoto, K. Detection of K-ras gene mutation by liquid biopsy in patients with pancreatic cancer. Cancer 2015, 121, 2271–2280. [Google Scholar] [CrossRef]
  31. Cancer Genome Atlas Research Network. Integrated Genomic Characterization of Pancreatic Ductal Adenocarcinoma. Cancer Cell 2017, 32, 185–203. [Google Scholar] [CrossRef]
  32. San Lucas, F.A.; Allenson, K.; Bernard, V.; Castillo, J.; Kim, D.U.; Ellis, K.; Ehli, E.A.; Davies, G.E.; Petersen, J.L.; Li, D.; et al. Minimally invasive genomic and transcriptomic profiling of visceral cancers by next-generation sequencing of circulating exosomes. Ann. Oncol. 2016, 27, 635–641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Allenson, K.; Castillo, J.; San Lucas, F.A.; Scelo, G.; Kim, D.U.; Bernard, V.; Davis, G.; Kumar, T.; Katz, M.; Overman, M.J.; et al. High prevalence of mutant KRAS in circulating exosome-derived DNA from early-stage pancreatic cancer patients. Ann. Oncol. 2017, 28, 741–747. [Google Scholar] [CrossRef]
  34. Erb, U.; Zoller, M. Progress and potential of exosome analysis for early pancreatic cancer detection. Expert Rev. Mol. Diagn. 2016, 16, 757–767. [Google Scholar] [CrossRef]
  35. Melo, S.A.; Luecke, L.B.; Kahlert, C.; Fernandez, A.F.; Gammon, S.T.; Kaye, J.; LeBleu, V.S.; Mittendorf, E.A.; Weitz, J.; Rahbari, N.; et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature 2015, 523, 177–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Lai, X.; Wang, M.; McElyea, S.D.; Sherman, S.; House, M.; Korc, M. A microRNA signature in circulating exosomes is superior to exosomal glypican-1 levels for diagnosing pancreatic cancer. Cancer Lett. 2017, 393, 86–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Exosomes Facilitate Therapeutic Targeting of Oncogenic KRAS in Pancreatic Cancer | Nature. Available online: https://www.nature.com/articles/nature22341?__hstc=12316075.07430159d50a3c91e72c280a7921bf0d.1525305600142.1525305600143.1525305600144.1&__hssc=12316075.1.1525305600145&__hsfp=1773666937 (accessed on 12 September 2020).
  38. Diefenbach, R.J.; Lee, J.H.; Kefford, R.F.; Rizos, H. Evaluation of commercial kits for purification of circulating free DNA. Cancer Genet 2018, 228–229, 21–27. [Google Scholar] [CrossRef] [PubMed]
  39. Ryan, D.P.; Hong, T.S.; Bardeesy, N. Pancreatic Adenocarcinoma. Available online: http://www.nejm.org/doi/10.1056/NEJMra1404198 (accessed on 15 September 2020).
  40. Takai, E.; Totoki, Y.; Nakamura, H.; Kato, M.; Shibata, T.; Yachida, S. Clinical Utility of Circulating Tumor DNA for Molecular Assessment and Precision Medicine in Pancreatic Cancer. Adv. Exp. Med. Biol. 2016, 924, 13–17. [Google Scholar] [CrossRef]
  41. Witkiewicz, A.K.; McMillan, E.A.; Balaji, U.; Baek, G.; Lin, W.-C.; Mansour, J.; Mollaee, M.; Wagner, K.-U.; Koduru, P.; Yopp, A.; et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat. Commun. 2015, 6, 6744. [Google Scholar] [CrossRef]
  42. Denis, J.A.; Guillerm, E.; Coulet, F.; Larsen, A.K.; Lacorte, J.-M. The Role of BEAMing and Digital PCR for Multiplexed Analysis in Molecular Oncology in the Era of Next-Generation Sequencing. Mol. Diagn. Ther. 2017, 21, 587–600. [Google Scholar] [CrossRef]
  43. Bennett, C.W.; Berchem, G.; Kim, Y.J.; El-Khoury, V. Cell-free DNA and next-generation sequencing in the service of personalized medicine for lung cancer. Oncotarget 2016, 7, 71013–71035. [Google Scholar] [CrossRef] [Green Version]
  44. Kyrochristos, I.D.; Ziogas, D.E.; Glantzounis, G.K.; Roukos, D.H. Prediction of pancreatic cancer risk and therapeutic response with next-generation sequencing. Biomark. Med. 2017, 12, 5–8. [Google Scholar] [CrossRef] [Green Version]
  45. Zill, O.A.; Greene, C.; Sebisanovic, D.; Siew, L.M.; Leng, J.; Vu, M.; Hendifar, A.E.; Wang, Z.; Atreya, C.E.; Kelley, R.K.; et al. Cell-Free DNA Next-Generation Sequencing in Pancreatobiliary Carcinomas. Cancer Discov. 2015, 5, 1040–1048. [Google Scholar] [CrossRef] [Green Version]
  46. Park, G.; Park, J.K.; Son, D.S.; Shin, S.H.; Kim, Y.J.; Jeon, H.J.; Lee, J.; Park, W.Y.; Lee, K.H.; Park, D. Utility of targeted deep sequencing for detecting circulating tumor DNA in pancreatic cancer patients. Sci. Rep. 2018, 8, 11631. [Google Scholar] [CrossRef]
  47. Liang, W.S.; Craig, D.W.; Carpten, J.; Borad, M.J.; Demeure, M.J.; Weiss, G.J.; Izatt, T.; Sinari, S.; Christoforides, A.; Aldrich, J.; et al. Genome-wide characterization of pancreatic adenocarcinoma patients using next generation sequencing. PLoS ONE 2012, 7, e43192. [Google Scholar] [CrossRef] [Green Version]
  48. Sausen, M.; Phallen, J.; Adleff, V.; Jones, S.; Leary, R.J.; Barrett, M.T.; Anagnostou, V.; Parpart-Li, S.; Murphy, D.; Kay Li, Q.; et al. Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients. Nat. Commun. 2015, 6, 7686. [Google Scholar] [CrossRef] [PubMed]
  49. Contreras-Naranjo, J.C.; Wu, H.-J.; Ugaz, V.M. Microfluidics for exosome isolation and analysis: Enabling liquid biopsy for personalized medicine. Lab Chip 2017, 17, 3558–3577. [Google Scholar] [CrossRef]
  50. Ludwig, N.; Whiteside, T.L.; Reichert, T.E. Challenges in Exosome Isolation and Analysis in Health and Disease. Int. J. Mol. Sci. 2019, 20, 4684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Zhu, Y.; Zhang, H.; Chen, N.; Hao, J.; Jin, H.; Ma, X. Diagnostic value of various liquid biopsy methods for pancreatic cancer: A systematic review and meta-analysis. Medicine (Baltimore) 2020, 99, e18581. [Google Scholar] [CrossRef]
  52. Sefrioui, D.; Blanchard, F.; Toure, E.; Basile, P.; Beaussire, L.; Dolfus, C.; Perdrix, A.; Paresy, M.; Antonietti, M.; Iwanicki-Caron, I.; et al. Diagnostic value of CA19.9, circulating tumour DNA and circulating tumour cells in patients with solid pancreatic tumours. Br. J. Cancer 2017, 117, 1017–1025. [Google Scholar] [CrossRef] [PubMed]
  53. Berger, A.W.; Schwerdel, D.; Costa, I.G.; Hackert, T.; Strobel, O.; Lam, S.; Barth, T.F.; Schröppel, B.; Meining, A.; Büchler, M.W.; et al. Detection of Hot-Spot Mutations in Circulating Cell-Free DNA From Patients With Intraductal Papillary Mucinous Neoplasms of the Pancreas. Gastroenterology 2016, 151, 267–270. [Google Scholar] [CrossRef]
  54. Liggett, T.; Melnikov, A.; Yi, Q.L.; Replogle, C.; Brand, R.; Kaul, K.; Talamonti, M.; Abrams, R.A.; Levenson, V. Differential methylation of cell-free circulating DNA among patients with pancreatic cancer versus chronic pancreatitis. Cancer 2010, 116, 1674–1680. [Google Scholar] [CrossRef] [PubMed]
  55. Castells, A.; Puig, P.; Móra, J.; Boadas, J.; Boix, L.; Urgell, E.; Solé, M.; Capellà, G.; Lluís, F.; Fernández-Cruz, L.; et al. K-ras mutations in DNA extracted from the plasma of patients with pancreatic carcinoma: Diagnostic utility and prognostic significance. J. Clin. Oncol. 1999, 17, 578–584. [Google Scholar] [CrossRef] [PubMed]
  56. Eissa, M.A.L.; Lerner, L.; Abdelfatah, E.; Shankar, N.; Canner, J.K.; Hasan, N.M.; Yaghoobi, V.; Huang, B.; Kerner, Z.; Takaesu, F.; et al. Promoter methylation of ADAMTS1 and BNC1 as potential biomarkers for early detection of pancreatic cancer in blood. Clin. Epigenet. 2019, 11, 59. [Google Scholar] [CrossRef] [PubMed]
  57. Maire, F.; Micard, S.; Hammel, P.; Voitot, H.; Lévy, P.; Cugnenc, P.-H.; Ruszniewski, P.; Puig, P.L. Differential diagnosis between chronic pancreatitis and pancreatic cancer: Value of the detection of KRAS2 mutations in circulating DNA. Br. J. Cancer 2002, 87, 551–554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Cohen, J.D.; Javed, A.A.; Thoburn, C.; Wong, F.; Tie, J.; Gibbs, P.; Schmidt, C.M.; Yip-Schneider, M.T.; Allen, P.J.; Schattner, M.; et al. Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers. Proc. Natl. Acad. Sci. USA 2017, 114, 10202–10207. [Google Scholar] [CrossRef] [Green Version]
  59. Campos-Carrillo, A.; Weitzel, J.N.; Sahoo, P.; Rockne, R.; Mokhnatkin, J.V.; Murtaza, M.; Gray, S.W.; Goetz, L.; Goel, A.; Schork, N.; et al. Circulating tumor DNA as an early cancer detection tool. Pharmacol. Ther. 2020, 207, 107458. [Google Scholar] [CrossRef]
  60. Kato, S.; Honda, K. Use of Biomarkers and Imaging for Early Detection of Pancreatic Cancer. Cancers 2020, 12, 1965. [Google Scholar] [CrossRef]
  61. Henriksen, S.D.; Madsen, P.H.; Larsen, A.C.; Johansen, M.B.; Drewes, A.M.; Pedersen, I.S.; Krarup, H.; Thorlacius-Ussing, O. Cell-free DNA promoter hypermethylation in plasma as a diagnostic marker for pancreatic adenocarcinoma. Clin. Epigenet. 2016, 8, 117. [Google Scholar] [CrossRef] [Green Version]
  62. Shen, S.Y.; Singhania, R.; Fehringer, G.; Chakravarthy, A.; Roehrl, M.H.A.; Chadwick, D.; Zuzarte, P.C.; Borgida, A.; Wang, T.T.; Li, T.; et al. Sensitive tumour detection and classification using plasma cell-free DNA methylomes. Nature 2018, 563, 579–583. [Google Scholar] [CrossRef]
  63. Poruk, K.E.; Gay, D.Z.; Brown, K.; Mulvihill, J.D.; Boucher, K.M.; Scaife, C.L.; Firpo, M.A.; Mulvihill, S.J. The Clinical Utility of CA 19-9 in Pancreatic Adenocarcinoma: Diagnostic and Prognostic Updates. Curr. Mol. Med. 2013, 13, 340–351. [Google Scholar]
  64. Kim, M.K.; Woo, S.M.; Park, B.; Yoon, K.-A.; Kim, Y.-H.; Joo, J.; Lee, W.J.; Han, S.-S.; Park, S.-J.; Kong, S.-Y. Prognostic Implications of Multiplex Detection of KRAS Mutations in Cell-Free DNA from Patients with Pancreatic Ductal Adenocarcinoma. Clin. Chem. 2018, 64, 726–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Singh, N.; Gupta, S.; Pandey, R.M.; Chauhan, S.S.; Saraya, A. High levels of cell-free circulating nucleic acids in pancreatic cancer are associated with vascular encasement, metastasis and poor survival. Cancer Investig. 2015, 33, 78–85. [Google Scholar] [CrossRef] [PubMed]
  66. Buscail, E.; Maulat, C.; Muscari, F.; Chiche, L.; Cordelier, P.; Dabernat, S.; Alix-Panabieres, C.; Buscail, L. Liquid Biopsy Approach for Pancreatic Ductal Adenocarcinoma. Cancers 2019, 11, 852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Earl, J.; Garcia-Nieto, S.; Martinez-Avila, J.C.; Montans, J.; Sanjuanbenito, A.; Rodríguez-Garrote, M.; Lisa, E.; Mendía, E.; Lobo, E.; Malats, N.; et al. Circulating tumor cells (CTC) and KRAS mutant circulating free DNA (cfDNA) detection in peripheral blood as biomarkers in patients diagnosed with exocrine pancreatic cancer. BMC Cancer 2015, 15, 797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Ako, S.; Nouso, K.; Kinugasa, H.; Dohi, C.; Matushita, H.; Mizukawa, S.; Muro, S.; Akimoto, Y.; Uchida, D.; Tomoda, T.; et al. Utility of serum DNA as a marker for KRAS mutations in pancreatic cancer tissue. Pancreatology 2017, 17, 285–290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Hadano, N.; Murakami, Y.; Uemura, K.; Hashimoto, Y.; Kondo, N.; Nakagawa, N.; Sueda, T.; Hiyama, E. Prognostic value of circulating tumour DNA in patients undergoing curative resection for pancreatic cancer. Br. J. Cancer 2016, 115, 59–65. [Google Scholar] [CrossRef]
  70. Vietsch, E.E.; van Eijck, C.H.; Wellstein, A. Circulating DNA and Micro-RNA in Patients with Pancreatic Cancer. Pancreat. Disord. Ther. 2015, 5. [Google Scholar] [CrossRef] [Green Version]
  71. Pishvaian, M.J.; Joseph Bender, R.; Matrisian, L.M.; Rahib, L.; Hendifar, A.; Hoos, W.A.; Mikhail, S.; Chung, V.; Picozzi, V.; Heartwell, C.; et al. A pilot study evaluating concordance between blood-based and patient-matched tumor molecular testing within pancreatic cancer patients participating in the Know Your Tumor (KYT) initiative. Oncotarget 2016, 8, 83446–83456. [Google Scholar] [CrossRef] [Green Version]
  72. Shu, Y.; Wu, X.; Tong, X.; Wang, X.; Chang, Z.; Mao, Y.; Chen, X.; Sun, J.; Wang, Z.; Hong, Z.; et al. Circulating Tumor DNA Mutation Profiling by Targeted Next Generation Sequencing Provides Guidance for Personalized Treatments in Multiple Cancer Types. Sci. Rep. 2017, 7, 583. [Google Scholar] [CrossRef]
  73. Bitterman, D.S.; Price, K.S.; Van Seventer, E.E.; Clark, J.W.; Allen, J.N.; Blaszkowsky, L.S.; Ryan, D.P.; Eyler, C.E.; Wo, J.Y.-L.; Hong, T.S.; et al. Noninvasive comprehensive genomic profiling from plasma ctDNA in pancreatic cancer patients. JCO 2020, 38, 753. [Google Scholar] [CrossRef]
  74. Cheng, H.; Liu, C.; Jiang, J.; Luo, G.; Lu, Y.; Jin, K.; Guo, M.; Zhang, Z.; Xu, J.; Liu, L.; et al. Analysis of ctDNA to predict prognosis and monitor treatment responses in metastatic pancreatic cancer patients. Int. J. Cancer 2017, 140, 2344–2350. [Google Scholar] [CrossRef] [PubMed]
  75. Glazer, E.S.; Rashid, O.M.; Klapman, J.B.; Harris, C.L.; Hodul, P.J.; Pimiento, J.M.; Malafa, M.P. Endoscopic ultrasonography complements computed tomography in predicting portal or superior mesenteric vein resection in patients with borderline resectable pancreatic carcinoma. Pancreatology 2017, 17, 130–134. [Google Scholar] [CrossRef] [PubMed]
  76. Sinn, M.; Bahra, M.; Liersch, T.; Gellert, K.; Messmann, H.; Bechstein, W.; Waldschmidt, D.; Jacobasch, L.; Wilhelm, M.; Rau, B.M.; et al. CONKO-005: Adjuvant Chemotherapy With Gemcitabine Plus Erlotinib Versus Gemcitabine Alone in Patients After R0 Resection of Pancreatic Cancer: A Multicenter Randomized Phase III Trial. J. Clin. Oncol. 2017, 35, 3330–3337. [Google Scholar] [CrossRef] [PubMed]
  77. Oettle, H.; Neuhaus, P.; Hochhaus, A.; Hartmann, J.T.; Gellert, K.; Ridwelski, K.; Niedergethmann, M.; Zulke, C.; Fahlke, J.; Arning, M.B.; et al. Adjuvant chemotherapy with gemcitabine and long-term outcomes among patients with resected pancreatic cancer: The CONKO-001 randomized trial. JAMA 2013, 310, 1473–1481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Sugiura, T.; Uesaka, K.; Kanemoto, H.; Mizuno, T.; Sasaki, K.; Furukawa, H.; Matsunaga, K.; Maeda, A. Serum CA19-9 is a significant predictor among preoperative parameters for early recurrence after resection of pancreatic adenocarcinoma. J. Gastrointest. Surg. 2012, 16, 977–985. [Google Scholar] [CrossRef]
  79. Fong, Z.V.; Alvino, D.M.L.; Fernandez-Del Castillo, C.; Mehtsun, W.T.; Pergolini, I.; Warshaw, A.L.; Chang, D.C.; Lillemoe, K.D.; Ferrone, C.R. Reappraisal of Staging Laparoscopy for Patients with Pancreatic Adenocarcinoma: A Contemporary Analysis of 1001 Patients. Ann. Surg. Oncol. 2017, 24, 3203–3211. [Google Scholar] [CrossRef]
  80. Adamo, P.; Cowley, C.M.; Neal, C.P.; Mistry, V.; Page, K.; Dennison, A.R.; Isherwood, J.; Hastings, R.; Luo, J.; Moore, D.A.; et al. Profiling tumour heterogeneity through circulating tumour DNA in patients with pancreatic cancer. Oncotarget 2017, 8, 87221–87233. [Google Scholar] [CrossRef] [Green Version]
  81. Brychta, N.; Krahn, T.; von Ahsen, O. Detection of KRAS Mutations in Circulating Tumor DNA by Digital PCR in Early Stages of Pancreatic Cancer. Clin. Chem. 2016, 62, 1482–1491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Almoguera, C.; Shibata, D.; Forrester, K.; Martin, J.; Arnheim, N.; Perucho, M. Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell 1988, 53, 549–554. [Google Scholar] [CrossRef] [Green Version]
  83. Takai, E.; Totoki, Y.; Nakamura, H.; Morizane, C.; Nara, S.; Hama, N.; Suzuki, M.; Furukawa, E.; Kato, M.; Hayashi, H.; et al. Clinical utility of circulating tumor DNA for molecular assessment in pancreatic cancer. Sci. Rep. 2015, 5, 18425. [Google Scholar] [CrossRef] [Green Version]
  84. Nishio, K.; Kimura, K.; Amano, R.; Yamazoe, S.; Ohrira, G.; Nakata, B.; Hirakawa, K.; Ohira, M. Preoperative predictors for early recurrence of resectable pancreatic cancer. World J. Surg. Oncol. 2017, 15, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Lee, B.; Lipton, L.; Cohen, J.; Tie, J.; Javed, A.A.; Li, L.; Goldstein, D.; Burge, M.; Cooray, P.; Nagrial, A.; et al. Circulating tumor DNA as a potential marker of adjuvant chemotherapy benefit following surgery for localized pancreatic cancer. Ann. Oncol. 2019, 30, 1472–1478. [Google Scholar] [CrossRef] [PubMed]
  86. McDuff, S.; Parikh, A.R.; Hazar-Rethinam, M.; Zheng, H.; Van Seventer, E.; Nadres, B.; Ryan, D.P.; Weekes, C.D.; Clark, J.W.; Fernandez-del Castillo, C.; et al. Using circulating tumor DNA (ctDNA) to predict surgical outcome after neoadjuvant chemoradiation for locally advanced pancreatic cancer (LAPC). JCO 2018, 36, 272. [Google Scholar] [CrossRef]
  87. Lee, J.-S.; Rhee, T.-M.; Pietrasz, D.; Bachet, J.-B.; Laurent-Puig, P.; Kong, S.-Y.; Takai, E.; Yachida, S.; Shibata, T.; Lee, J.W.; et al. Circulating tumor DNA as a prognostic indicator in resectable pancreatic ductal adenocarcinoma: A systematic review and meta-analysis. Sci. Rep. 2019, 9, 16971. [Google Scholar] [CrossRef] [PubMed]
  88. Pietrasz, D.; Pécuchet, N.; Garlan, F.; Didelot, A.; Dubreuil, O.; Doat, S.; Imbert-Bismut, F.; Karoui, M.; Vaillant, J.-C.; Taly, V.; et al. Plasma Circulating Tumor DNA in Pancreatic Cancer Patients Is a Prognostic Marker. Clin. Cancer Res. 2017, 23, 116–123. [Google Scholar] [CrossRef] [Green Version]
  89. Nakano, Y.; Kitago, M.; Matsuda, S.; Nakamura, Y.; Fujita, Y.; Imai, S.; Shinoda, M.; Yagi, H.; Abe, Y.; Hibi, T.; et al. KRAS mutations in cell-free DNA from preoperative and postoperative sera as a pancreatic cancer marker: A retrospective study. Br. J. Cancer 2018, 118, 662–669. [Google Scholar] [CrossRef] [Green Version]
  90. Groot, V.P.; Mosier, S.; Javed, A.A.; Teinor, J.A.; Gemenetzis, G.; Ding, D.; Haley, L.M.; Yu, J.; Burkhart, R.A.; Hasanain, A.; et al. Circulating Tumor DNA as a Clinical Test in Resected Pancreatic Cancer. Clin. Cancer Res. 2019, 25, 4973–4984. [Google Scholar] [CrossRef]
  91. Tie, J.; Kinde, I.; Wang, Y.; Wong, H.L.; Roebert, J.; Christie, M.; Tacey, M.; Wong, R.; Singh, M.; Karapetis, C.S.; et al. Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer. Ann. Oncol. 2015, 26, 1715–1722. [Google Scholar] [CrossRef]
  92. Lee, B.; Cohen, J.; Lipton, L.R.; Tie, J.; Javed, A.A.; Li, L.; Goldstein, D.; Cooray, P.; Nagrial, A.; Burge, M.E.; et al. Potential role of circulating tumor DNA (ctDNA) in the early diagnosis and post-operative management of localised pancreatic cancer. JCO 2017, 35, 4101. [Google Scholar] [CrossRef]
  93. Azizian, A.; Rühlmann, F.; Krause, T.; Bernhardt, M.; Jo, P.; König, A.; Kleiß, M.; Leha, A.; Ghadimi, M.; Gaedcke, J. CA19-9 for detecting recurrence of pancreatic cancer. Sci. Rep. 2020, 10, 1332. [Google Scholar] [CrossRef] [Green Version]
  94. Däbritz, J.; Preston, R.; Hänfler, J.; Oettle, H. Follow-Up Study of K-ras Mutations in the Plasma of Patients With Pancreatic Cancer: Correlation With Clinical Features and Carbohydrate Antigen 19-9. Pancreas 2009, 38, 534–541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Del Re, M.; Vivaldi, C.; Rofi, E.; Vasile, E.; Miccoli, M.; Caparello, C.; d’Arienzo, P.D.; Fornaro, L.; Falcone, A.; Danesi, R. Early changes in plasma DNA levels of mutant KRAS as a sensitive marker of response to chemotherapy in pancreatic cancer. Sci. Rep. 2017, 7, 7931. [Google Scholar] [CrossRef] [PubMed]
  96. Bernard, V.; Kim, D.U.; San Lucas, F.A.; Castillo, J.; Allenson, K.; Mulu, F.C.; Stephens, B.M.; Huang, J.; Semaan, A.; Guerrero, P.A.; et al. Circulating Nucleic Acids Are Associated With Outcomes of Patients With Pancreatic Cancer. Gastroenterology 2019, 156, 108–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Kim, S.T.; Lim, D.H.; Jang, K.-T.; Lim, T.; Lee, J.; Choi, Y.-L.; Jang, H.-L.; Yi, J.H.; Baek, K.K.; Park, S.H.; et al. Impact of KRAS Mutations on Clinical Outcomes in Pancreatic Cancer Patients Treated with First-line Gemcitabine-Based Chemotherapy. Mol. Cancer Ther. 2011, 10, 1993–1999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Semrad, T.; Barzi, A.; Lenz, H.-J.; Hutchins, I.M.; Kim, E.J.; Gong, I.-Y.; Tanaka, M.; Beckett, L.; Holland, W.; Burich, R.A.; et al. Pharmacodynamic separation of gemcitabine and erlotinib in locally advanced or metastatic pancreatic cancer: Therapeutic and biomarker results. Int. J. Clin. Oncol. 2015, 20, 518–524. [Google Scholar] [CrossRef] [Green Version]
  99. Nagai, M.; Sho, M.; Akahori, T.; Nakagawa, K.; Nakamura, K. Application of liquid biopsy for surgical management of pancreatic cancer. Ann. Gastroenterol. Surg. 2020, 4, 216–223. [Google Scholar] [CrossRef] [Green Version]
  100. Xu, Y.F.; Hannafon, B.N.; Zhao, Y.D.; Postier, R.G.; Ding, W.Q. Plasma exosome miR-196a and miR-1246 are potential indicators of localized pancreatic cancer. Oncotarget 2017, 8, 77028–77040. [Google Scholar] [CrossRef]
  101. Yang, S.; Che, S.P.Y.; Kurywchak, P.; Tavormina, J.L.; Gansmo, L.B.; de Sampaio, P.C.; Tachezy, M.; Bockhorn, M.; Gebauer, F.; Haltom, A.R.; et al. Detection of mutant KRAS and TP53 DNA in circulating exosomes from healthy individuals and patients with pancreatic cancer. Cancer Biol. Ther. 2017, 18, 158–165. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Potential impact of ctDNA-based liquid biopsy on multimodal management of pancreatic ductal adenocarcinoma (PDAC). Created with BioRender (BioRender.com, accessed 10 September 2020). CT, computed tomography.
Figure 1. Potential impact of ctDNA-based liquid biopsy on multimodal management of pancreatic ductal adenocarcinoma (PDAC). Created with BioRender (BioRender.com, accessed 10 September 2020). CT, computed tomography.
Ijms 21 07651 g001
Figure 2. Sample collection, storage, DNA isolation, amplification and sequencing, and clinical application are important steps of ctDNA liquid biopsy. Created with BioRender (BioRender.com, accessed 10 September 2020). PCR, polymerase chain reaction.
Figure 2. Sample collection, storage, DNA isolation, amplification and sequencing, and clinical application are important steps of ctDNA liquid biopsy. Created with BioRender (BioRender.com, accessed 10 September 2020). PCR, polymerase chain reaction.
Ijms 21 07651 g002
Table 1. Current trials registered with clinicaltrials.gov exploring liquid biopsy with cfDNA in pancreatic cancer.
Table 1. Current trials registered with clinicaltrials.gov exploring liquid biopsy with cfDNA in pancreatic cancer.
Trial NumberTrial NamectDNA Liquid Biopsy Focus/GoalsStudy TypeEstimated Completion Recruitment Status
NCT02079363DNA Promoter Hypermethylation as a Blood Based Maker for Pancreatic CancerAssessment of hypermethylation as diagnostic, prognostic, and recurrence markerProspective Observational CohortJanuary 2018Unknown
NCT03524677Mutation of K-RAS, CDKN2A, SMAD4 and TP53 in Pancreatic Cancer: Role of Liquid Biopsy in Preoperative DiagnosisAssessment of four ctDNA mutations’ impact on preoperative staging and progressionProspective Observational CohortJanuary 2020Recruiting
NCT02934984Circulating Cell-free Tumor DNA (ctDNA) in Pancreatic CancerctDNA as a tool for surveillance/screening for recurrenceProspective Observational CohortJanuary 2021Recruiting
NCT04246203Prognostic Role of Circulating Tumor DNA in Resectable Pancreatic Cancer (PROJECTION)ctDNA as prognostic indicator in patients with radiographically resectable cancerProspective Observational CohortMarch 2025Not yet recruiting
NCT04241367Verification of Predictive Biomarkers for Pancreatic Cancer Treatment Using Multicenter Liquid BiopsyAssessment of ctDNA KRAS mutation on outcomes in pancreas cancer. Will also assess ctDNA for specific gene targetsProspective Observational CohortDecember 2025Recruiting
NCT04176952PRIMUS002: Looking at 2 Neo-adjuvant Treatment Regimens for Resectable and Borderline Resectable Pancreatic CancerctDNA liquid biopsy as a means to stratify response to a chemotherapy regimen (secondary outcome) Phase 2 Clinical TrialDecember 2023Recruiting
NCT04484636PLATON - Platform for Analyzing Targetable Tumor Mutations (Pilot-study) (PLATON)Assessing peripheral blood samples for targetable mutations in multiple gastrointestinal malignanciesProspective Observational CohortJune 2021Not yet recruiting
NCT03334708A Study of Blood Based Biomarkers for Pancreas AdenocarcinomaDetermination of sensitivity and specificity of ctDNA for the diagnosis of early stage pancreatic cancerProspective Observational CohortOctober 2021Recruiting
NCT03568630Blood Markers of Early Pancreas CancerIdentification of blood markers of early pancreas cancerProspective Observational CohortJuly 2023Recruiting
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Grunvald, M.W.; Jacobson, R.A.; Kuzel, T.M.; Pappas, S.G.; Masood, A. Current Status of Circulating Tumor DNA Liquid Biopsy in Pancreatic Cancer. Int. J. Mol. Sci. 2020, 21, 7651. https://doi.org/10.3390/ijms21207651

AMA Style

Grunvald MW, Jacobson RA, Kuzel TM, Pappas SG, Masood A. Current Status of Circulating Tumor DNA Liquid Biopsy in Pancreatic Cancer. International Journal of Molecular Sciences. 2020; 21(20):7651. https://doi.org/10.3390/ijms21207651

Chicago/Turabian Style

Grunvald, Miles W., Richard A. Jacobson, Timothy M. Kuzel, Sam G. Pappas, and Ashiq Masood. 2020. "Current Status of Circulating Tumor DNA Liquid Biopsy in Pancreatic Cancer" International Journal of Molecular Sciences 21, no. 20: 7651. https://doi.org/10.3390/ijms21207651

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

Article Metrics

Back to TopTop