Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2246 2024-02-16 19:50:02 |
2 layout + 29 word(s) 2275 2024-02-26 09:46:59 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Ramírez-Maldonado, E.; López Gordo, S.; Major Branco, R.P.; Pavel, M.; Estalella, L.; Llàcer-Millán, E.; Guerrero, M.A.; López-Gordo, E.; Memba, R.; Jorba, R. Clinical Application of Liquid Biopsy in Pancreatic Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/55097 (accessed on 02 May 2024).
Ramírez-Maldonado E, López Gordo S, Major Branco RP, Pavel M, Estalella L, Llàcer-Millán E, et al. Clinical Application of Liquid Biopsy in Pancreatic Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/55097. Accessed May 02, 2024.
Ramírez-Maldonado, Elena, Sandra López Gordo, Rui Pedro Major Branco, Mihai-Calin Pavel, Laia Estalella, Erik Llàcer-Millán, María Alejandra Guerrero, Estrella López-Gordo, Robert Memba, Rosa Jorba. "Clinical Application of Liquid Biopsy in Pancreatic Cancer" Encyclopedia, https://encyclopedia.pub/entry/55097 (accessed May 02, 2024).
Ramírez-Maldonado, E., López Gordo, S., Major Branco, R.P., Pavel, M., Estalella, L., Llàcer-Millán, E., Guerrero, M.A., López-Gordo, E., Memba, R., & Jorba, R. (2024, February 16). Clinical Application of Liquid Biopsy in Pancreatic Cancer. In Encyclopedia. https://encyclopedia.pub/entry/55097
Ramírez-Maldonado, Elena, et al. "Clinical Application of Liquid Biopsy in Pancreatic Cancer." Encyclopedia. Web. 16 February, 2024.
Clinical Application of Liquid Biopsy in Pancreatic Cancer
Edit

Pancreatic ductal adenocarcinoma contributes significantly to global cancer-related deaths, featuring only a 10% survival rate over five years. The quest for novel tumor markers is critical to facilitate early diagnosis and tailor treatment strategies for this disease, which is key to improving patient outcomes. In pancreatic ductal adenocarcinoma, these markers have been demonstrated to play a crucial role in early identification, continuous monitoring, and prediction of its prognosis and have led to better patient outcomes. Nowadays, biopsy specimens serve to ascertain diagnosis and determine tumor type. However, liquid biopsies present distinct advantages over conventional biopsy techniques. They offer a noninvasive, easily administered procedure, delivering insights into the tumor’s status and facilitating real-time monitoring. Liquid biopsies encompass a variety of elements, such as circulating tumor cells, circulating tumor DNA, extracellular vesicles, microRNAs, circulating RNA, tumor platelets, and tumor endothelial cells.

Pancreatic ductal adenocarcinoma liquid biopsy circulating tumor cells circulating tumor DNA circulating free tumor DNA

1. Impact of Liquid Biopsy in Early and Differential Diagnosis of Pancreatic Ductal Adenocarcinoma 

The challenge in the clinical practice in this pathology (pancreatic intraepithelial neoplasia (PanIN) or early stage PDAC) lies in the early detection to improve the prognosis of PDAC.  Liquid biopsy (LB) holds the potential to enhance detection without subjecting the patient to the risks associated with aggressive diagnostic tools [1]. A systematic review and meta-analysis from Zhu and colleagues described the studies performed on LB methods in detecting PDAC [2]. The sensitivity and specificity of LB were 80% and 89%, respectively, with an area under the curve (AUC) of 0.936 [1][2] (Figure 1 and Table 1).
The analysis of various body fluids, including pancreatic juice, saliva and urine, has been investigated for the early detection of PDAC. Mutations in Kristen rat sarcoma (KRAS) have been detected in the pancreatic juice and in biliary cytobrush specimens, where selective detection has been observed in PDAC patients over benign lesions [3]. Mutation levels of KRAS2 were higher in patients with PDAC compared to chronic pancreatitis (CP). In CP, telomerase activity and alterations in genes such as CDKN2A/p16, TP53 and SMAD4/DPC4 could aid in PDAC diagnosis [4][5]. The analysis of biomarkers in saliva offers an easy and noninvasive way to search for mutations in RNAs and miRNAs of genes such as MBD3L2, KRAS, ACRV1 and DPMI, among others, which could assist in differentiating pancreatic cancer patients from CP patients and healthy control [6]. Other biomarkers in RNAs and miRNAs have been described [7][8]. Analysis of genetic mutations in urine could also facilitate detection of PDAC [9][10].
Importantly, the efficacy of LB to detect PDAC varies depending on the tumor stage and biomarker type. For example, circulating tumor DNA (ctDNA) is less frequently detected in patients with resectable disease compared to those with unresectable disease [1]. Here, researchers outline the utility of various biomarkers in detecting PDAC within the general population and among specific risk groups (Figure 1 and Table 1).
Figure 1. Clinical application of liquid biopsy in pancreatic ductal adenocarcinoma [11]. LB: Liquid biopsy; NAC: Neoadjuvant chemotherapy.
Table 1. Comparison between biomarkers of pancreatic ductal adenocarcinoma [12][13][14].
  ctDNA Circulating Tumor Cells (CTCs) Extracellular Vesicles (EVs)
Target KRAS, TP53, CDKN2A, SMAD4, BRAF, PIK3CA, ADAMTS1, BNC1, 5MC, H2AZ, H2A1.1, H3K4me2, h2ak119ub CD45, CEP8, CK, EpCAM KRAS, TP53, RNA: miRNA, longRNA
Proteins markers: EFGR, EPCAM, MUC-1, GPC-1, WNT2
Isolation Blood Blood Body fluids
Tumor information Epigenetic information DNA, RNA, Protein DNA, RNA, Protein
Technological approaches qPCR, dPCR, ddPCR, NGS, commercial kits Immunoaffinity, Physical methods (size and density) Density-based, size-based, affinity-based, commercial kits
Advantages qPCR: Fast and low-cost
dPCR: High sensitivity/Specificity
NGS: capability to screen for a broad range of genetic variants using high DNA input
Immunoaffinity: Specific, label-free obtained
Physical methods: Fast, simple, Low-cost, label-free obtained
Density-based: low cost. Independent of marker expression.
Size-based: Low-cost, fast, Independent of marker expression.
Affinity-based: Specificity. High purity.
Commercial kits: Simple, fast.
Disadvantages General: No early stages
qPCR: Low sensitivity. Only points mutations.
dPCR: High cost. Only points mutations.
NGS: Variable sensitivity. High cost.
General: Isolation complex and expensive. Technical variability
Immunoaffinity: capture only one subpopulation. Low purity.
Physical methods: Needs immuno-labeling techniques to distinguish CTCs
General: Isolation complex by contamination and expensive
Density-based: Time, high volume sample, can damage EVs.
Size based: contamination.
Affinity-based: low sample yield.
Commercial kits: High cost.
Sensitivity (S) (%) 34–71%
KRAS mutations: codons 12, 13, 61, in different stages.
73–76%
CD45/CEP8
100% Mt, 58% resectable
Anti-EpCAM portal vein Blood
67% ES, 80% LA, 85% Mt
KRAS mutations in exoDNA
50% ES
GPC1
miRNAs
Increased expression
Specificity (Sp) (%) 75–81%
Mutations KRAS exon 2
68%
CD45/CEP8
90% ES
GPC1
Combined techniques (%) S: 85–98%, Sp: 77–81%
ctDNA (KRAS exon 2) with CA19.9
S: 47%
ctDNA (KRAS MAFs) with CA19.9
S: 100%, Sp: 80%
CTCs.with EVs
NR
Application No suitable for screening of PDAC
Monitoring postoperative minimal residual disease
Predictor of disease recurrence and prognosis
Not present in healthy controls
Variable sensitivity in early diagnosis
Excellent specificity.
Follow-up of disease recurrence and prognosis
Functional analysis drug resistance
The highest sensitivity and specificity in early detection
Evaluated response of resection or any therapy
Biotherapeutic application
BEAMing: beads-emulsion-amplification-magnetics; CEP8: chromosome 8 centromere; ctDNA: circulating tumor DNA; CTC: circulating tumor cells; dPCR: digital polymerase chain reaction; ddPCR: droplet digital PCR; EpCAM: epithelial cell adhesion molecule; ES: early stages; EVs: extracellular vesicles; GPC1: glypican-1; LA: locally advanced; miRNA: micro-RNA; Mt: metastatic; NGS: next-generation sequencing; NR: not reported.

1.1. Circulating Tumor DNA

Some studies found that cfDNA is detected at higher levels from PDAC patients compared to pancreatic neuroendocrine tumors or CP patient [15] and have been associated with poor disease-specific survival [15][16][17]. Of all mutated genes detected from ctDNA in PDAC patients, KRAS is the one most frequently found (50–90%). Although mutations can also be found in healthy controls and patients with CP, its mutation levels are significantly higher in PDAC [16][18].
In their review, Zhu and colleagues emphasized that although the sensitivity of ctDNA is marginally lower than that of CTCs, ctDNA provides considerably higher specificity [2]. Notably, while the detection of ctDNA is deemed appropriate for the diagnosis of PDAC, it is not considered suitable for screening purposes. The limited sensitivity of ctDNA in early-stage PDAC is attributed to minimal cellular necrosis at this stage, resulting in the release of only a small quantity of ctDNA into the peripheral bloodstream [2].

1.2. Circulating Tumor Cells

Pancreatic cells can indeed be identified in the bloodstream even before tumor development. In contrast to healthy individuals, these cells are detectable in 33% of patients with cystic lesions [19] and in 73% of those with PDAC [19][20]. Furthermore, other studies have noted the presence of CTCs in varying proportions in benign, premalignant, or malignant lesions, but not in healthy controls [21].
The diagnostic efficacy of CTCs, however, has been a topic of debate among researchers due to their inconsistent sensitivity, which ranges from 21% to 100% [22]. In a study by Ankeni and colleagues, a NanoVelcro CTCs microfluidic chip was employed to analyze 100 sequential samples from pretreatment PDAC patients. The results showed CTCs in 54 out of 72 confirmed PDAC patients, demonstrating a sensitivity of 75% and a specificity of 96.4% when used for diagnostic purposes [23].
Given the technical variability in detecting CTCs, some researchers recommend combining the detection of CTCs with other biomarkers to enhance sensitivity up to 100% and specificity to 80% [24]. Although the specificity of CTCs is lower than that of EVs, their utilization still presents considerable diagnostic potential in PDAC detection [2].

1.3. Extracellular Vesicles

EVs from pancreatic cells are easily detectable in peripheral blood owing to their abundant levels, a consequence of the exocrine function of these cells, and possess a longer half-life compared to ctDNA. Interestingly, KRAS mutations in ctDNA were found in 7.4% and 14.8% of healthy donors when tested from exosomal DNA (exoDNA) or cfDNA, respectively [25], and in 13% of CP patients [26]. Research by Melo et al. demonstrated that glypican-1 (GPC1) levels in EVs were significantly higher in PDAC patients compared to those with benign pancreatic diseases or healthy individuals, showcasing remarkable sensitivity and specificity (100%) [27]. Moreover, studies by Zhang et al. [28] and Lewis et al. [29] employed ‘chips’ to detect GPC1, aiming to enhance sensitivity and specificity in distinguishing PDAC from healthy samples. Other studies have shown that combining EVs analysis with CA19-9 could improve specificity [30]. Notably, EVs analysis proves useful for both diagnosing and screening PDAC, offering superior diagnostic value compared to other techniques, partly due to its high AUC of 0.9819 [2]. To summarize, among various detection methods, EVs exhibit the highest diagnostic efficacy, sensitivity, and AUC [2].

1.4. miRNAs

miRNAs are noncoding, single-stranded RNA molecules up to 22 nucleotides long that function as posttranscriptional gene expression regulators [31]. Various miRNAs have been utilized to differentiate PDAC patients from those with benign lesions, CP, and healthy controls, and a JAMA-published study reported 38 significantly dysregulated miRNAs in PDAC patients compared to controls [32]. These include miR-223, miR-23b-3p, miR-100, miR-205, miR-192-5p, a six-miRNA panel, miR-483-3p, miR-99 (a and b), miR-21, miR-25, and miR-205, among others. The detection of diverse miRNAs could enhance diagnostic accuracy and distinguish PDAC from healthy individuals [33]. In pancreatic conditions such as intraductal papillary mucinous neoplasm [34] or pancreatic intraepithelial neoplasia [35], miRNAs have been instrumental in differentiating patients with high-grade dysplasia or early-stage PDAC from healthy controls. Importantly, a meta-analysis revealed that miRNAs offer a sensitivity of 79% and specificity of 74% for early PDAC diagnosis. Similar to other biomarkers, combining analysis of miRNAs with CA19-9 analysis improved the quality of data, yielding an AUC of 0.84 [36]. Moreover, whilst miRNA analysis from pancreatic juice demonstrated a specificity of 88% and sensitivity of 87%, the inclusion of serum CA19-9 levels enhanced sensitivity to 91% and specificity to 100% [37]. In saliva samples, hsa-miR-21, hsa-miR-23a, hsa-miR-23b, and miR-29c were significantly upregulated in PDAC patients compared to controls, reporting sensitivities of 71.4%, 85.7%, 85.7%, and 57%, respectively, and a specificity of 100% [38].
Numerous studies exploring biomarker combinations in healthy individuals, CP, and PDAC patients [39][40] have produced various results, with a common trend pointing towards higher specificity when combining biomarkers. For instance, one study demonstrated that using at least two biomarkers among CA19-9, CTCs, or ctDNA achieved a sensitivity and specificity of approximately 80% and 90%, respectively [41][42]. Another study confirmed that combining CTCs with CA19-9 elevated the positive diagnostic rate for PDAC to 97.5% [43].

2. Role of Liquid Biopsy after Resection of Pancreatic Ductal Adenocarcinoma

Surgery stands as the singular potentially curative intervention for PDAC, given the tumor’s notable resistance to chemotherapy, radiation, and immunotherapy. Attaining an R0 resection represents the optimal opportunity for patients to achieve a favorable prognosis, resulting in an enhanced 5-year survival rate ranging from 8% to 25% [1][2][12][13][14][18][44][45]. Despite undergoing curative resection, disease recurrence significantly impacts postoperative outcomes, with over 70% of resected PDAC patients succumbing to recurrent disease [1][2][12][13][14][18]. Consequently, there is a critical need for an effective strategy to identify minimal residual disease (R1) during or postsurgery and to anticipate the risk of recurrence [45]. LB emerges as a promising approach to monitor disease progression in PDAC following surgical intervention [1][2][12][13][14][18][44][45] (Figure 1 and Table 1).

2.1. Circulating Tumor DNA

Despite the initial hypothesis suggesting that tumor components are released into the bloodstream following manipulation, a recent meta-analysis revealed that surgical resection of resectable primary tumors plays a role in the negativization of ctDNA [45]. However, this comprehensive analysis failed to establish a significant impact of ctDNA negativization on the overall survival (OS) and disease-free survival (DFS) rates of PDAC patients. The authors emphasized the considerable heterogeneity observed within the analyzed studies, attributing it to variations in postoperative determinations of ctDNA, which extended beyond 24 h after the removal of surgical specimens. Considering the relatively short half-life of ctDNA, ranging from minutes to hours, delayed determinations increase the likelihood of encountering false-negative results. On the other hand, the elevation in ctDNA levels induced by surgical trauma can persist for 2 to 4 weeks, potentially concealing persistent ctDNA in patients experiencing relapse. Consequently, the authors recommended conducting LB between 2 and 4 weeks postsurgery to minimize the risk of false negatives [1][2][12][13][14][18].
Recent studies have indicated that the detection of ctDNA in preoperative blood samples from PDAC patients may identify candidates for NAC. Additionally, elevated ctDNA levels were associated with an increased risk of death, and patients with a LB after surgery showing as positive for ctDNA exhibited a higher recurrence rate [45]. The persistence of ctDNA positivity postsurgical resection is predictive of a shorter DFS, possibly indicating the presence of occult micrometastases or residual local disease, supporting the consideration of additional adjuvant treatment [14][18][44][45].

2.2. Circulating Tumor Cells 

In a recent randomized trial, the authors implemented a no-touch pancreaticoduodenectomy, involving the manipulation of the tumor only after the complete isolation of vascular and lymphatic drainage vessels [46]. The study reported a reduction in CTCs in the portal vein when employing the no-touch technique but no discernible improvement in OS compared to standard surgery. In contrast, a meta-analysis conducted by Vidal and colleagues failed to demonstrate a decreased release of tumoral components after surgical manipulation employing the no-touch technique, and no superior OS and DFS were observed compared to standard surgery [45].
Importantly, the presence of CTCs has been shown to progressively increase in advanced diseases. For instance, a study revealed that patients with occult metastases of PDAC exhibited significantly higher CTCs counts than those without metastatic disease. Therefore, considering CTC levels becomes crucial in understanding the disease state and thus determining whether NAC should be favored over surgical treatment [44]. Additionally, a prospective study assessed CTC levels in preoperative PDAC patients, establishing a direct correlation between elevated CTC levels and disease recurrence at one year in patients undergoing resection [14].

2.3. Extracellular Vesicles 

Several studies have suggested that EVs derived from tumors can serve as indicators of the response to surgery and treatment, offering a potential avenue for a reliable marker in PDAC. Similar to other markers, the levels of miRNAs originating from EVs return to normal within 24 h of PDAC resection. However, persistently elevated levels postsurgery may indicate the presence of hidden metastasis. Such cases warrant vigilant follow-up, and individuals may require additional treatment following surgery [44]. A study investigating miR-451a, miR-4525, and miR-21 from EVs obtained from portal venous blood during pancreatectomy revealed elevated levels emerging as an independent prognostic factor for OS and DFS [14].

References

  1. Heredia-Soto, V.; Rodríguez-Salas, N.; Feliu, J. Liquid biopsy in pancreatic cancer: Are we ready to apply it in the clinical practice? Cancers 2021, 13, 1986.
  2. 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 2020, 99, e18581.
  3. Yang, J.; Li, S.; Li, J.; Wang, F.; Chen, K.; Zheng, Y.; Wang, J.; Lu, W.; Zhou, Y.; Yin, Q.; et al. A meta-analysis of the diagnostic value of detecting K-ras mutation in pancreatic juice as a molecular marker for pancreatic cancer. Pancratology 2016, 16, 605–614.
  4. Shi, C.; Fukushima, N.; Abe, T.; Bian, Y.; Hua, L.; Wendelburg, B.J.; Yeo, C.J.; Hruban, R.H.; Goggins, M.G.; Eshleman, J.R. Sensitive and quantitative detection of KRAS2 gene mutations in pancreatic duct juice differentiates patients with pancreatic cancer from chronic pancreatitis, potential for early detection. Cancer Biol. Ther. 2008, 7, 353–360.
  5. Hata, T.; Ishida, M.; Motoi, F.; Yamaguchi, T.; Naitoh, T.; Katayose, Y.; Egawa, S.; Unno, M. Telomerase activity in pancreatic juice differentiates pancreatic cancer from chronic pancreatitis: A meta-analysis. Pancreatology 2016, 16, 372–381.
  6. Zhang, L.; Farrell, J.J.; Zhou, H.; Elashowff, D.; Akin, D.; Park, N.H.; Chia, D.; Wong, D.T. Salivary transcriptomic biomarkers for detection of resectable pancreatic cancer. Gastroenterology 2010, 138, 949–957.
  7. Xie, Z.; Chen, X.; Li, J.; Guo, Y.; Li, H.; Pan, X.; Jiang, J.; Liu, H.; Wu, B. Salivary HOTAIR and PVT1 as novel biomarkers for early pancreatic cancer. Oncotarget 2016, 7, 25408–25419.
  8. Xie, Z.; Yin, X.; Gong, B.; Nice, W.; Wu, B.; Zhang, X.; Huang, J.; Zhang, P.; Zhou, Z.; Li, Z. Salivary microRNAs show potential as a noninvasive biomarker for detecting resectable pancreatic cancer. Cancer Prev. Res. 2015, 8, 165–173.
  9. Yoshizawa, N.; Sugimoto, K.; Tameda, M.; Inagaki, Y.; Ikejiri, M.; Inoue, H.; Usui, M.; Takei, Y. miR-3940-5p/miR-8069 ratio in urine exosomes is a novel diagnostic biomarker for pancreatic ductal adenocarcinoma. Once Lett. 2020, 19, 2677–2684.
  10. Terasawa, H.; Kinugasa, H.; Ako, S.; Hirai, M.; Matsushita, H.; Uchida, D.; Tomoda, T.; Matsumoto, K.; Horiguchi, S.; Kato, H.; et al. Utility of liquid biopsy using urine in patients with pancreatic ductal adenocarcinoma. Cancer Biol. Ther. 2019, 20, 1348–1353.
  11. Tempero, M.A.; Malafa, M.P.; Al-Hawari, M.; Behrman, S.W.; Benson, A.B.; Cardin, D.B.; Chiorean, E.G.; Chung, V.; Czito, B.; Del Chiaro, M.; et al. Pancreatic adenocarcinoma, Version 1.2024, NCCN Clinical practice guidelines in oncology. J. Natl. Cancer Netw. 2021, 19, 430–457.
  12. Watanabe, F.; Suzuki, K.; Noda, H.; Rikiyama, T. Liquid biopsy leads to a paradigm shift in the treatment of pancreatic cancer. World J. Gastroenterol. 2022, 28, 6478–6496.
  13. Rofi, E.; Vivaldi, C.; Del Re, M.; Arrigoni, E.; Crucitta, S.; Fuel, N.; Fogli, S.; Vasile, E.; Musettini, G.; Fornaro, L.; et al. The emerginn role of liquid biopsy in diagnosis, prognosis, an treatment monitoring of pancreatic cancer. Pharmacogenomics 2019, 20, 49–68.
  14. Raufi, A.G.; May, M.S.; Hadfield, M.J.; Seyhan, A.A.; El-Deiry, W.S. Advances in liquid biopsy technology and implications for pancreatic cancer. Int. J. Mol. Sci. 2023, 24, 4238.
  15. Adamo, P.; Cowley, C.M.; Neal, C.P.; Mistry, V.; Page, K.; Denninson, 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.
  16. Finkelstein, S.D.; Bibbo, M.; Loren, D.E.; Siddiqui, A.A.; Solomides, C.; Kowalski, T.E.; Ellsworth, E. Molecular analysis of centrifugation supernatant fluid from pancreaticobiliary duct samples can improve cancer detection. Acta Cytol. 2012, 56, 439–447.
  17. Sikora, K.; Bedin, C.; Vicentini, C.; Malpeli, G.; D’Angelo, E.; Sperandio, N.; Lawlor, R.T.; Bassi, C.; Tortora, G.; Nitti, D.; et al. Evaluation of cell-free DNA as a biomarker for pancreatic malignancies. Int. J. Biol. Markers 2015, 30, e136–e141.
  18. 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.
  19. Rhim, A.D.; Thege, F.I.; Santana, S.M.; Lannin, T.B.; Saha, T.N.; Tsai, S.; Maggs, L.R.; Kochman, M.L.; Gingsberg, G.G.; Lieb, J.G.; et al. Detection of circulating pancreas epithelial cells in patients with pancreatic cystic lesions. Gastroenterology 2014, 146, 647–651.
  20. Kulemann, B.; Pitman, M.B.; Liss, A.S.; Valsangkar, N.; Fernández-Del Castillo, C.; Lillemoe, K.D.; Hoeppner, J.; Mino-Kenudson, M.; Warshaw, A.L.; Thayer, S.P. Circulating tumor cells found in patients with localized and advanced pancreatic cancer. Pancreas 2015, 44, 547–550.
  21. Cauley, C.E.; Pitman, M.B.; Zhou, J.; Perkins, J.; Coleman, B.; Liss, A.S.; Fernández-Del Castillo, C.; Warshaw, A.L.; Lillemoe, K.D.; Ayer, S.P. Circulating Epithelial Cells in Patients with Pancreatic Lesions: Clinical and Pathologic Findings. J. Am. Coll. Surg. 2015, 221, 699–707.
  22. Qi, Z.H.; Xu, H.X.; Zhang, S.R.; Xu, J.Z.; Li, S.; Gao, H.L.; Jin, W.; Wang, W.Q.; Wu, C.T.; Ni, Q.X.; et al. The Significance of Liquid Biopsy in Pancreatic Cancer. J. Cancer 2018, 9, 3417–3426.
  23. Ankeny, J.S.; Court, C.M.; Hou, S.; Li, Q.; Song, M.; Wu, D.; Chen, J.F.; Lee, T.; Lin, M.; Sho, S.; et al. Circulating tumour cells as a biomarker for diagnosis and staging in pancreatic cancer. Br. J. Cancer 2016, 114, 1367–1375.
  24. Buscail, E.; Alix-Panabieres, C.; Quincy, P.; Cauvin, T.; Chauvet, A.; Degrandi, O.; Caumont, C.; Verdom, S.; Lamrissi, I.; Moranvillier, I.; et al. 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 2019, 11, 1656.
  25. Allenson, K.; Castillo, J.; San Lucas, F.A.; Scale, 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.
  26. 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.
  27. Melo, S.A.; Luecke, L.B.; Kahlert, C.; Fernandez, A.F.; Gaming, 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.
  28. Zhang, J.; Zhu, Y.; Shi, J.; Zhang, K.; Zhang, Z.; Zhang, H. Sensitive Signal Amplifying a Diagnostic Biochip Based on a Biomimetic Periodic Nanostructure for Detecting Cancer Exosomes. ACS Appl. Mater. Interfaces 2020, 12, 33473–33482.
  29. Lewis, J.M.; Vyas, A.D.; Qiu, Y.; Messer, K.S.; White, R.; Heller, M.J. Integrated Analysis of Exosomal Protein Biomarkers on Alternating Current Electrokinetic Chips Enables Rapid Detection of Pancreatic Cancer in Patient Blood. ACS Nano 2018, 12, 3311–3320.
  30. Lux, A.; Kahlert, C.; Grützmann, R.; Pilarsky, C. c-Met and PD-l1 on circulating exosomes as diagnostic and prognostic markers for pancreatic cancer. Int. J. Mol. Sci. 2019, 20, 3305.
  31. Yan, T.B.; Huang, J.Q.; Huang, S.Y.; Ahir, B.K.; Li, L.M.; Mo, Z.N.; Zhong, J.H. Advances in the Detection of Pancreatic Cancer Through Liquid Biopsy. Front. Oncol. 2021, 11, 801173.
  32. Schultz, N.A.; Dehlendorff, C.; Jensen, B.V.; Bjerregaard, J.K.; Nielsen, K.P.; Bojesen, S.E.; Calatayud, D.; Nielsen, S.E.; Yilmaz, M.; Holländer, N.H.; et al. MicroRNA biomarkers in whole blood for detection of pancreatic cancer. JAMA 2014, 311, 392–404.
  33. Yan, Q.; Hu, D.; Li, M.; Chen, Y.; Wu, X.; Ye, Q.; Wang, Z.; He, L.; Zhu, J. The Serum MicroRNA Signatures for Pancreatic Cancer Detection and Operability Evaluation. Front. Bioeng. Biotechnol. 2020, 8, 379.
  34. Komatsu, S.; Ichikawa, D.; Miyamae, M.; Kawaguchi, T.; Morimura, R.; Hirajima, S.; Okajima, W.; Ohashi, T.; Imamura, T.; Konishi, H.; et al. Malignant potential in pancreatic neoplasm; New insights provided by circulating miR-223 in plasma. Expert Opin. Biol. Ther. 2015, 15, 773–785.
  35. Bartsch, D.K.; Gercke, N.; Strauch, K.; Wieboldt, R.; Matthäi, E.; Wagner, V.; Rospleszcz, S.; Schäfer, A.; Franke, F.S.; Mintziras, I.; et al. The combination of miRNA-196b, LCN2, and TIMP1 is a potential set of circulating biomarkers for screening individuals at risk for familial pancreatic cancer. J. Clin. Med. 2018, 7, 295.
  36. 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.
  37. Wang, J.; Raimondo, M.; Guha, S.; Chen, J.; Diao, L.; Dong, X.; Wallacew, M.B.; Killary, A.M.; Frazier, M.L.; Woodward, T.A.; et al. Circulating microRNAs in pancreatic juice as candidate biomarkers of pancreatic cancer. J. Cancer 2014, 5, 696–705.
  38. Humeau, M.; Vignolle-Vidoni, A.; Sicard, F.; Martins, F.; Burnt, B.; Buscail, L.; Torrisani, J.; Cordelier, P. Salivary microRNA in pancreatic cancer patients. PLoS ONE 2015, 10, e0130996.
  39. Xiao, D.; Dong, Z.; Zhen, L.; Xia, G.; Huang, X.; Wang, T.; Geo, H.; Yang, B.; Xu, C.; Wu, W.; et al. Combined exosomal GPC1, CD82, and serum CA19-9 as multiplex targets: A specific, sensitive, and reproducible detection panel for the diagnosis of pancreaticcancer. Mol. Cancer Res. 2020, 18, 1300–1310.
  40. Pu, X.; Ding, G.; Wu, M.; Zhou, S.; Jia, S.; Cao, L. Elevated expression of exosomal microRNA–21 as a potential biomarker for the early diagnosis of pancreatic cancer using a tethered cationic lipoplex nanoparticle biochip. Oncol. Lett. 2020, 19, 2062–2070.
  41. Sefrioui, D.; Blanchard, F.; Toure, E.; Basile, P.; Beaussire, L.; Dolfus, C.; Perdix, A.; Pares, 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.
  42. Wu, H.; Guo, S.; Liu, X.; Li, Y.; Su, Z.; He, Q.; Liu, X.; Zhang, Z.; Yu, L.; Shi, X.; et al. Noninvasive detection of pancreatic ductal adenocarcinoma using the methylation signature of circulating tumour DNA. BMC Med. 2022, 20, 458.
  43. 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, 622.
  44. 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.
  45. Vidal, L.; Pando, E.; Blanco, L.; Fabregat-Franco, C.; Castet, F.; Sierra, A.; Macarulla, T.; Balsells, J.; Charco, R.; Vivancos, A. Liquid biopsy after resection of pancreatic adenocarcinoma and its relation to oncological outcomes. Systematic review and meta-analysis. Cancer Treat. Rev. 2023, 120, 102604.
  46. Leon, S.A.; Shapiro, B.; Sklaroff, D.M.; Yaros, M.J. Free DNA in the Serum of Cancer Patients and the Effect of Therapy. Cancer Res. 1977, 37, 646–650.
More
Information
Subjects: Surgery
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , , ,
View Times: 68
Revisions: 2 times (View History)
Update Date: 26 Feb 2024
1000/1000