Review Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Apr 21, 2023; 29(15): 2241-2260
Published online Apr 21, 2023. doi: 10.3748/wjg.v29.i15.2241
Emerging role of non-invasive and liquid biopsy biomarkers in pancreatic cancer
Akash Bararia, Nilabja Sikdar, Human Genetics Unit, Indian Statistical Institute, Kolkata 700108, India
Prosenjeet Chakraborty, Department of Molecular Biosciences, SVYASA School of Yoga and Naturopathy, Bangalore 560105, India
Paromita Roy, Department of Pathology, Tata Medical Center, Kolkata 700160, India
Bitan Kumar Chattopadhay, Department of General Surgery, IPGMER & SSKM Hospital, Kolkata 700020, India
Amlan Das, Department of Biochemistry, Royal Global University, Assam 781035, India
Aniruddha Chatterjee, Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9061, New Zealand
Aniruddha Chatterjee, School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun 248007, India
ORCID number: Nilabja Sikdar (0000-0003-4465-472X).
Author contributions: Bararia A and Chakraborty P wrote the first draft of the paper and constructed the flow chart and tables; Chatterjee A, Roy P, Chattopadhay B K, Das A and Sikdar N provided valuable input, suggestions, comments and guidance while writing the review manuscript and contributed to proofreading and editing; Sikdar N constructed, wrote, conceptualized, and edited the review manuscript.
Supported by the Department of Biotechnology, Government of India Grant Sanction, Ramalingaswami Re-entry Fellowship, No. RLS/BT/Re-entry/05/2012.
Conflict-of-interest statement: The authors declare no conflicts of interest.
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: Nilabja Sikdar, PhD, Research Scientist, Doctor, Human Genetics Unit, Indian Statistical Institute, 203, B.T. Road, Kolkata 700108, India. snilabja@isical.ac.in
Received: November 28, 2022
Peer-review started: November 28, 2022
First decision: January 11, 2023
Revised: February 2, 2023
Accepted: March 15, 2023
Article in press: March 15, 2023
Published online: April 21, 2023
Processing time: 136 Days and 23.7 Hours

Abstract

A global increase in the incidence of pancreatic cancer (PanCa) presents a major concern and health burden. The traditional tissue-based diagnostic techniques provided a major way forward for molecular diagnostics; however, they face limitations based on diagnosis-associated difficulties and concerns surrounding tissue availability in the clinical setting. Late disease development with asymptomatic behavior is a drawback in the case of existing diagnostic procedures. The capability of cell free markers in discriminating PanCa from autoimmune pancreatitis and chronic pancreatitis along with other precancerous lesions can be a boon to clinicians. Early-stage diagnosis of PanCa can be achieved only if these biomarkers specifically discriminate the non-carcinogenic disease stage from malignancy with respect to tumor stages. In this review, we comprehensively described the non-invasive disease detection approaches and why these approaches are gaining popularity for their early-stage diagnostic capability and associated clinical feasibility.

Key Words: Non-invasive biomarkers, Cell free biomarkers, Proteomic biomarkers, Liquid biopsy-based diagnostics, Pancreatic cancer biomarkers

Core Tip: Considering the limited commercial availability of cell free nucleic acids and secreted proteome-based non-invasive biomarkers it is crucial to summarize them for the proper diagnosis of pancreatic cancer and distinguish it from other benign pancreatic diseases. We also highlighted the clinical use of these non-invasive biomarkers in diagnostics. This review will successfully guide readers to address the current issues and aid in the cutting-edge development of biomarkers with higher sensitivity and specificity.



INTRODUCTION

The global population is severely burdened by the increase in multiple types of cancer. Cancer-related incidence and its associated mortality have emerged as a major threat to global health in developed countries, like the United States, European countries, Canada, and Australia, and developing as well as under-developed countries including India and Southeast Asian countries. Pancreatic cancer (PanCa) is a deadly and emerging type of cancer because of its associated poor survival. Only 20% of patients are eligible for surgery due to the physiologically deep-rooted location and the generalized symptoms caused by the close proximity to crucial vessels. In 2022, the estimated new incidences in the United States alone are 62210 (male) and 32970 (female), and the estimated deaths associated with PanCa are 25970 males and 23860 females.

Per the GLOBACON 2020 data, PanCa is the 12th most common cause of cancer with 495000 new cases across the globe with approximately 47% of the new cases registered in Asia and another major proportion of 28% in Europe. There might be an increase in the incidence by 70% by the end of 2022, which translates into approximately 844000 new cases per year. As documented in the United States population, PanCa-related mortality has relatively increased in men than in women. In the United Kingdom, PanCa is the tenth most frequent cancer type, and it ranks fifth in terms of cancer-related deaths per year. Some risk factors associated with PanCa apart from genetic predisposition include obesity, diet and smoking. PanCa-associated surgery has a high mortality risk making it a more life-threatening disease[1-4]. Based on the demography and ethnicity of patients, the profile of biomarkers and predisposition to particular risk factors can vary[4,5].

LIQUID BIOPSY AND ITS ROLE IN PANCA DIAGNOSIS

Liquid biopsy (also known as a fluid biopsy) is an approach to identify markers released from tumors in the blood or associated body fluids. It includes but is not limited to cell-free DNA (cfDNA), cell-free RNA (cfRNA), cell-free proteins, circulating tumor cells and exosomes[6-8]. Liquid biopsy is rapidly developing and has a highly emerging market with an estimated $5 billion (USD) turnover globally by 2023[9]. DNA is the most dominant approach in terms of biomarker identification from plasma or serum and in terms of availability for clinical practices, which makes cfDNA a lucrative option.

There are multiple factors that make liquid biopsy an attractive option in comparison to tissue biopsy. A major reason is that traditional tissue biopsy fails to identify tissue heterogeneity in depth whereas liquid biopsy does that more efficiently[10]. The reason behind this is based on the concept that various categories of tumor materials are released in the blood irrespective of tumor heterogeneity. Therefore, making the liquid biopsy samples, such as plasma or serum, a cocktail that has tumor markers irrespective of cell type is not present in the case of the traditional biopsy technique. In addition, the exponential growth of next generation sequencing technologies enables the identification of cell free nucleic acids with high sensitivity, which is of great clinical application.

Moreover, traditional tissue biopsy is not applicable in the case of patients that are in an early stage and have not developed a tumor of significant size[11]. Early-stage PanCa is usually asymptomatic, and no operative options are available to examine the tumor, making traditional tissue biopsy non-functional in this context. For early or advanced diagnosis and disease assessment, liquid biopsy can be a major step forward toward cancer prevention and screening of populations. APAPTEST was the first cancer blood-based test done to identify markers in the case of cervical cancer[12]. Disease-targeted liquid biopsy-based biomarkers can detect patients who have an intermediate chance of developing PanCa such as those having chronic pancreatitis (CP), diabetes or a genetic predisposition to PanCa. It can also distinguish between PanCa and pancreatic diseases that are immune system modulated in their origin.

A crucial requirement is distinguishing autoimmune pancreatitis (AIP) from PanCa. AIP diagnosis, which is an autoimmune disease based on the rise in IgG4 levels in serum, is based on a liquid biopsy-based approach for identification and differentiation from malignancies such as PanCa. Upregulation of IgG4-positive plasma cells in organ tissues followed by increased IgG4 levels in the serum is usually defined as type 1 AIP. On the contrary, type 2 AIP has no connection with IgG4. It demonstrates features in the pancreatic ducts resembling granulocytic epithelial lesions. Type 1 AIP also acts as a pancreatic lesion related to IgG4-related disease[13]. Since both type I AIP and type II AIP are based on identification using non-invasive approaches, the development of these techniques is urgent to support cases where invasive approaches are a less viable option. This approach acts as a powerful alternative to existing diagnostic methods and will be highly beneficial for the identification of cases with aggressive tumor biology and support clinicians to make proper individualized targeted therapy[13-17].

The methylation status alteration in cancer cells is also an important point to consider for disease diagnosis. The nature of methylation based alterations discriminates itself from genetic changes based on tumor tissue type specificity, which is crucial in the case of methylation alterations apart from being more pervasive and prevalent in terms of occurrences[18,19]. Aberrant DNA methylation alterations mostly occur in CpG islands and play a vital role in activating tumor suppressor genes and oncogenes, thereby modulating the genome[20-23]. The approach to identifying cfDNA-based methylation changes is a growing area of interest because invasive approaches are not always feasible and proactive in terms of clinical diagnostics[24,25]. Various reports claimed that in the case of pancreatic ductal adenocarcinoma (PDAC) an altered methylation signature has been observed when tumor tissues were compared with normal tissues, hence directing its implications in biomarker development[26,27].

The neoplastic tendency of the pancreas needs to be identified, and specific non-invasive markers discriminating PanCa from CP or other pancreatic precursor lesions such as pancreatic intraepithelial neoplasia (PanIN), intrapapillary mucinous neoplasia (IPMN) and mucinous cystic neoplasm need to be developed[28]. Luo et al[29] documented that cg10673833, a circulating tumor DNA (ctDNA) methylation marker was found to be very sensitive (89.7%) and specific (86.8%) for the detection of colorectal cancer and its associated pre-cancerous lesions. SEPT9 was the first blood-based screening test that the Food and Drug Administration approved for colorectal cancer. Cancer-seek was developed by Tivey et al[30]. It is a blood test that detects circulatory protein and cfDNA biomarkers. It is capable of detecting eight cancer types, mostly in their respective advanced stages, with a specificity of 99% and a sensitivity of 69%-98%, which varied based on cancer type[12,20].

PANCA AND ITS ASSOCIATED BENIGN DISEASES

Systemic approaches to develop biomarkers to distinguish PDAC from other precancerous lesions are of great importance as they will also support the early diagnosis of PDAC and substantially improve outcomes. Single gene analysis has its limitation in early detection because it has been observed that the specificity and sensitivity of biomarkers in combination increase more than single biomarkers. Traditional serum-based biomarkers like CA19-9, CA125, and carcinoembryonic antigen (CEA) for PanCa diagnostics are of immense importance, but there is a need for the development of more sensitive and specific non-invasive biomarkers. The sensitivity and specificity of detection are usually much higher when the above three biomarkers are used jointly instead of individually[21,31]. In addition, varied combinations of DNA methylation-based biomarkers are being developed to better understand the complexity of the diseases including both the initiation and neoplastic stages. Concurrently addressing the contribution of immune cells in the cfDNA pool and its associated methylation or hydroxyl-methylation pattern is of great importance as it can be obtained from plasma samples[32-35].

Almost 50 of 100000 people are affected by CP worldwide with an increasing trend, and a CP-associated prolonged inflammatory state enhances tissue conversion to neoplasia. Pancreatic tumor (PT) formation is also increasing along with CP worldwide. Mortality due to PTs is close to 6%. After a diagnosis of CP, it takes almost 10 years before tumor symptoms appear, which is a long latency period with sufficient time for early diagnosis and treatment via non-invasive approaches. PENK DNA methylation has been observed broadly in precancerous lesions of various grades including extraductal and intraductal PTs and CP along with PanIN, IPMN and mucinous cystic neoplasm. PENK methylation alterations increased as the neoplastic tissue grade increased but was absent in the case of AIP and adjacent normal pancreatic tissue[36].

In a study of an 87 gene panel, 6 genes, namely PRKC, IKZF1, CD1D, KCNK12, CLEC11A and NDRG4, were shown to have clinical ability and potential as a cfDNA diagnostic biomarker for PanCa[37]. In this study using pancreatic juice taken from PanCa, CP and healthy patients, the authors documented that most of the biomarkers were successful in discriminating patients with PanCa from healthy patients, but only CD1D was successful in discriminating patients with CP from PanCa with a sensitivity of 84%[37,38]. A remarkable change in methylation status was documented by Liggett et al[39], where they showcased that hypomethylated genes in CP converted to hypermethylated genes in PanCa, suggesting that altered methylation is also a driving reason for neoplasia development in the case of PTs.

A total of 14 genes, namely CCND2, CDKN2B, DAPK1, MUC2, MYOD1, promoter A of MLH1, CDKN1C, ESR1, MGMT PGK1, the proximal region of the PGR promoter, RARB, RB1 and SYK, were successful in distinguishing CP from PanCa based on cfDNA DNA methylation profiling with 91% sensitivity and specificity[38,39]. In a study to identify cell death specific to tissue type by Lehmann-Werman et al[40], they observed that CUX2 and REG1A are specific to pancreas tissue type. They also found these genes to be hypomethylated in plasma in both PanCa and CP, but the levels were lowered in other lower neoplastic grade lesions. CUX2 was more hypomethylated in the case of PanCa and REG1A was higher in CP, which gives the ability to discriminate CP from PanCa based on tissue death irrespective of etiology[38,40].

In a study using pancreatic juice samples collected from PanCa and other associated precancerous lesions, six genes were demonstrated to be differentially methylated between CP and PanCa[41]. The six genes, TFPI2, NPTX2, FOXE1, p16, ppENK and CyclinD2, were able to successfully distinguish between CP and PanCa as CP had lower levels of methylation than PanCa. Similarly, the levels differed amongst healthy tissue and high-risk patients creating a gradient of altered methylation across various stages starting from healthy to precancerous to PanCa samples. It was also able to distinguish between IPMN and PanCa based on cfDNA methylation from pancreatic juice samples. Further, the study also attempted to address the limitations of IPMN diagnostics, which cytology mostly fails[42]. Promoter DNA methylation of the CDO1 gene was identified to be involved in early-stage PanCa based on cytological experiments. cfDNA methylation analysis of CDO1 from PanCa, CP and AIP patients showed that significant promoter methylation was associated with PanCa but was absent in CP and AIP[43]. In another study, CD1D showed cfDNA methylation when analyzed in pancreatic juice samples of PanCa [area under the curve (AUC): 0.92], but no methylation was documented in benign diseases[21,43]. In a study by Humeau et al[44], they reported that urine and sputum are also suitable alternatives to plasma for cell-free nucleic acid biomarker development. The utility of other material such as urine as a source of non-invasive testing needs further data and investigation.

MicroRNA (miRNA), namely hsa-miR-23a and hsa-miR-23b, have been documented to be overexpressed in IPMN patient saliva, which makes it a suitable precancerous lesion biomarker. It was also observed that hsa-miR-210 along with let-7c signals may be leveraged from saliva in distinguishing CP from healthy subjects[32,44]. Su et al[45] concurrently conducted a study using serum as a sample source and observed that miR-877, miR-3201, miR-890, miR-602 and miR-16-2-3p were successful in distinguishing low-risk and high-risk PDAC[32,45]. Irrespective of this development, there is still a hindrance in using miRNAs in clinical diagnostics due to reproducibility issues and variable expression of miRNAs. However, substantial work is ongoing in this area[32].

In a study to differentiate PDAC from CP by Bosch et al[46], it was documented that the the presence of a high level of soluble AXL (sAXL) was associated with lower overall survival in the case of PDAC. When identified from plasma, a higher level of sAXL was documented in PDAC than in CP and healthy controls, and they concluded that it is a potential biomarker for discriminating CP from PDAC and useful in identifying the onset of PDAC by its increasing level during monitoring. The findings were also validated in murine models. sAXL was also found to be overexpressed in IPMN, which is a pre-PDAC stage[46].

Weeks et al[47] conducted research where they studied the urine proteome differences amongst PDAC, CP and healthy individuals. They noticed 101 proteins that were significantly altered in PanCa and CP. S100A9 was found to be downregulated in PanCa but overexpressed in CP. Annexin A2, CD59 and gelsolin proteins were also found to be dysregulated in PDAC samples[47]. An isotope-coded affinity tag tended to successfully distinguish between PanCa and CP based on quantitative protein expression[41]. In this review, we documented the important cell free biomarkers that are vital in the early-stage detection of PanCa and are able to distinguish PanCa from its non-malignant precursors.

CFDNA AND ITS ASSOCIATED SECRETED PROTEOME
cfDNA in PanCa and its association with benign pancreatic diseases

First reported in 1948 by Mandel and Meta[48], cfDNA is undoubtedly the most dominant form among the liquid biopsy-based approaches and when released by tumor is also a ctDNA. Leon et al[49] in 1977 introduced cfDNA levels, which were found to be notably more advanced in malignancies than in healthy individuals thereby showcasing its role in carcinogenesis. Similar observations were made in 1983 with PDAC patients, and it was noted that cfDNA levels were higher in PDAC patients than in CP and acute pancreatitis[50]. The origin of cfDNA in the blood can be due to release from tumors, apoptosis or necrosis and other active cellular secretions. A major factor leading to the rise in cfDNA studies in cancer management is the half-life of cfDNA, which is around 16 min-2.5 h, and that it can be monitored for overall disease progression and associated clinical prognosis with medical treatment (Table 1)[12,49,51].

Table 1 Cell free DNA involved in pancreatic cancer diagnostic and prognostics.
Target candidate
Up/down
Sample
Potential value
CEA mRNAUpWhole bloodD
EGFR mRNAUpSerumD
4GnT mRNAUpWhole bloodD
COL6A3 mRNAUpSerumD/P
Mir-155UpPlasmaD
miR-196aUpPlasmaD
miR-21UpPlasmaD
miR-210UpPlasmaD
miR-155UpPancreatic juiceD
miR-21UpPlasmaD
miR-196aUpSerumD/P
miR-200aUpSerumD
miR-200bUpPlasmaD
miR-210UpPlasmaD
miR-18aUpPlasmaD/T
miR-16UpPlasmaD
miR-196aUpPlasmaD
miR-185UpSerumD
miR-191UpSerumD
miR-20aUpSerumD
miR-21UpSerumD/P
miR-24UpSerumD
miR-25UpSerumD
miR-99aUpSerumD
miR-1290UpSerumD
miR-221UpPlasmaD/T/P
miR-375DownPlasmaD/T/P
miR-375UpPlasmaD
miR-27a3pUpWhole bloodD
miR-196aUpSerumD
miR-196bUpSerumD
miR-205UpPancreatic juiceD/P
miR-210UpSalivaD/P
miR-492UpPlasmaD/P
miR-1427UpPlasmaD/P
miR-22UpPlasmaD
miR-642bUpPlasmaD
miR-885-5pUpPlasmaD
Multigene indexUpWhole bloodD
miR-483-3pUpPlasmaD
MIR-21UpSerumD
MIR-210DownSerumD
MIR-155UpSerumD
miR-222DownSerumD
miR-203DownSerumD
miR-132DownSerumD
miR-212DownSerumD
miR-96DownSerumD
mi-126DownSerumD
mi-217DownSerumD
Let-7DownSerumD
miR-144DownSerumD
miR-148DownSerumD
miR-34aDownSerumD
miR-3548UpSerumD

Cancer diagnosis and treatment face a major setback from recurrence of the malignancy that cannot be ideally monitored using traditional techniques due to the lack of proper tumor formation and its associated diagnostic techniques. Postoperative or post-chemotherapeutic analysis of ctDNA in the blood can also be referred to as minimal residual disease (MRD) analysis. Minimal residual disease analysis cannot be ideally performed using imaging and traditional biopsy techniques, and the analysis using cfDNA/ctDNA is a great advance to measure tumor recurrence (Table 1)[12,52].

In terms of aberrant DNA methylation, which is also a common observation in PDAC patients, it has been documented that a change in methylation levels has been observed in various stages of dysplasia[53,54]. This indicates that the level of cfDNA methylation across various pre-PanCa lesions to PDAC is altered providing a candidate opportunity to track PDAC progression. In 2009, Melnikov et al[55] established the use of cfDNA methylation in PDAC diagnosis from 30 plasma samples. This was completed using microarray technology where they identified that the promoter region was hypomethylated in five genes with a sensitivity and specificity of 76% and 59%, respectively[55]. A crucial step in PanCa treatment is staging as that provides a proper clinical treatment approach and is associated with DNA promoter hypermethylation. Henriksen et al[56] reported that the use of cfDNA promoter hypermethylation as a prognostic biomarker in PDAC staging, which is based on the hypothesis that cfDNA methylation is stage-specific, can help in stage classification. Henriksen et al[56] analyzed a set of 28 genes in PDAC plasma samples. They developed the first prognostic prediction model using cfDNA hypermethylation targeting PDAC staging. They separated cluster cases with and without distant metastasis. The researchers were also able to document significantly higher numbers of hypermethylated genes clustering in stage IV PDAC than in earlier stages amongst which BNC1 was the most significant gene of interest (Table 1)[25,27,56].

A crucial challenge in cell free nucleosome identification for the differentiation of benign pancreatic disease from PanCa is a nonspecific rise of cell free nucleosome levels in the serum of patients while quantifying the cell free nucleosome. Bauden et al[36] identified the cell free nucleosome epigenetic alterations specific to disease type and stages by isolating intact nucleosomes and then quantifying them using an enzyme-linked immunosorbent assay. They were successful in quantifying five serum cell free nucleosome diagnostic biomarkers with an AUC of 0.95, which differentiated PanCa from a healthy pancreas. The epigenetic profiles of cell free nucleosomes often differ between individual patient serum samples based on cancer and healthy populations. Understanding that the epigenetic changes usually occur at the onset of the neoplastic transformation stages, those already in pre-neoplastic stages may represent cell free nucleosome profiles that can act as possible cell free biomarkers for the early detection of cancer (Table 1)[36].

Role of cfDNA as a diagnostic and therapeutic biomarker

Tumors in the pancreas can occur in tissues that are hard to isolate due to their depth of location and also small tumor size can be a limitation. Therefore, in this context blood-based cell free biomarkers are of high clinical importance[56]. Melnikov et al[55] investigated that they developed the MethDet56 test which contained screening of 56 gene promoters from PDAC plasma samples that identified five promoters namely CCND2, SOCS1, THBS1, PLAU and VHL with significant aberrant DNA methylation and with sensitivity and specificity of 76% and 59% respectively. These are initial biomarker development work showcasing that blood-based biomarkers could be highly valuable in diagnosis and prognosis if further studied and developed[55]. In a study by Sato et al[57], three out of six genes, namely CLDN5, SFRP1 and NPTX2, previously validated in tissue samples were found to be differentially methylated in pancreatic juice samples collected from PTs[38,57]. However, Matsubayashi et al[42] concluded that five genes that were previously validated by solid biopsy failed were not replicated in liquid biopsy. Although the genes TFPI2, PENK, CCND2, NPTX2 and FOXE1 were successfully able to discriminate PanCa from healthy tissue, the analysis failed to discriminate CP from PanCa or healthy tissue based on cfDNA methylation (Table 1)[42,55].

Yi et al[58] interestingly found the use of BNC1 and ADAMTS1 as novel serum-based biomarkers for early PanCa detection. The same research group concluded that low frequencies of these two genes were observed in pancreatitis, and there was a significant methylation difference between PanINs and PanCa. BNC1 could still be detected in PanIN-1, but ADAMTS1 could only be detected significantly in invasive PanCas[58]. Li et al[59] reported in an analogous study that BNC1 and SEPT9 were found to be highly methylated in PanCa and were suggested to have a significant role in PanCa diagnostic cfDNA biomarker development from plasma. A limitation of the study was that cfDNA methylation level in plasma was also found to be elevated in other benign diseases such as CP, acute pancreatitis and other lesions as these are risk factors leading to PT formation. This suggests a possible role of methylation changes in driving the progression of benign lesions to metastatic PanCa[60].

Global epigenetic alterations in 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) work as standalone entities in carcinogenesis[61]. Cao et al[62] concluded in this context that 5mC as well as 5hmC work as effective cfDNA in PDAC diagnostics and that when 5mC-5hmC work as an integrated system it provides an enhanced diagnostic power. As reported by Guler et al[33], PanCa early detection is suitable using 5hmC-based detection techniques, and a regression model showed an AUC of 0.919 encompassing a highly variable gene set. The low cfDNA methylation observation in the case of PDAC was consistent with other studies, and it corresponded to KRAS mutation increase in malignancy. PDAC 5-hmC increase was observed at the transcription start site and 3’ untranslated region, but a decrease was seen in the promoters[33]. cfDNA-based KRAS mutation identification is usually a good indicator of neoplasia though with limited specificity. KRAS remains a widely used gene for the early stage detection of PanCa. Other gene mutations, such as DNMT3A, TP53, GNAS, JAK2 and BCORL1, remain alternative ctDNA-based biomarkers[24,63-65]. However, it is clear that analysis of somatic genes in ctDNA provides limited sensitivity in detecting early-stage tumors (Table 1).

Cell free protein (secreted proteome) in PanCa and its associated benign pancreatic diseases

The secreted proteome often acts as a suitable diagnostic biomarker, but currently only CA19-9 is a Food and Drug Administration-approved biomarker for PDAC diagnosis. It has substantial drawbacks and lacks a proper proteomic dataset for PanCa. Often false negative CA19-9 results are given by patients with FUT3 mutations, and false positives are also observed in non-malignant CP patients as CA19-9 is usually very highly overexpressed[46].

The plasma-free amino acids (PFAAs) index is growing in popularity as a suitable approach for the diagnosis of PanCa. A study to compare the PFAAs index between PanCa and healthy controls by Fukutake et al[66] found PFAAs as a significant biomarker capable of distinguishing PDAC. The receiver operating curve AUC developed index was demonstrated to suitably discriminate PanCa from controls. PFAAs are modulated by metabolic changes, in particular in organ systems based on disease induction. As a result, most metabolomic studies consider the analysis of the PFAAs index[66]. Sources of protein biomarker analysis are usually blood and pancreatic juice, but it can also be validated from tissue samples. Wu et al[21] documented that altered levels of proteins, namely GPC1, PFAA, OPNT + TIMP-1, CPA4, MUC5AC and C4BPA, were observed in the serum of PanCa patients. They also documented that GPC1 level was significantly higher in PanCa patients than in patients with benign pancreatic diseases such as CP or AIP. It was also documented that serum levels of cell free CPA4 were significantly higher in PanCa than in healthy controls with a parallel observation that C4BPA was comparatively more sensitive than CA19-9. A crucial observation was based on PFAA where it was revealed that PFAA levels correlated with PanCa staging and that can be used for pathological PanCa staging. A significantly high level of serine in plasma was also documented along with a significant decrease in threonine levels in PanCa patient plasma (Table 2)[21,66,67].

Table 2 Cell free protein (secreted proteome) involved in pancreatic cancer diagnostic and prognostics.
Target candidate
Up/down
Sample
Target
Pathway
Potential value
ADAMTS1UpPlasmaADAMTS1MethylationD
BNC1UpPlasmaBNC1MethylationD
CDKN1CUpPlasmaCDKN1CMethylationD
MLH1DownPlasmaMLH1MutationD
PGR (prox)UpPlasmaPGRCell signalingD
SYKUpPlasmaSYK protein, mRNACell signalingD
CCND2UpPlasmaCyclin D2Cell signalingD
ALX4UpPlasmaTranscription factorMutationP
APCDownPlasmaAPCMutationP
BMP3UpPlasmaTGFβCell signalingP
BNC1UpPlasmaBNC1Cell signalingP
BRCA1UpPlasmaBRCA1MutationP
CDKN2BUpPlasmaInk4, p16, p14Cell signalingP
CHFRUpPlasmaE3 Ubiquitin-proteinCell signaling, check pointP
ESR1UpPlasmaESR1, Transcription factorCell signalingP
EYA2UpPlasmaEYA2Cell cycleP
GSTP1UpPlasmaGSTP1Cell signalingP
HIC1UpPlasmaHIC ZBTB TRANSCRIPTIONAL RECEPTOR 1Cell signalingP
MEST1v2UpPlasmaAlpha/Beta hydrolase superfamilyCell signalingP
Foxe1DownPancreatic juiceFoxe1MethylationD
MGMTUpPlasmaMGMTMethylationP
MLH1UpPlasmaMLH1MutationP
NPTX2UpPancreatic juiceNP, synaptic proteinsCell signalingP
NEUROG1UpPlasmaNEUROG1Cell signaling, regulatory networkP
RARBUpPlasmaRARBCell signalingP
RASSF1ADownPlasmaRAS effector proteinMethylationP
SFRP2UpPlasmaSFRP2Wnt signalingP
SEPT9v2UpPlasmaSeptin9Cell cycleP
CDIDUpPancreatic juiceCDIDMethylationD
PRKCUpPancreatic juicePRKCMethylationD
IKZF1UpPancreatic juiceIKZF1MethylationD
KCNK12UpPancreatic juiceKCNK12MethylationD
CLEC11AUpPancreatic juiceCLEC11AMethylationD/P
NDRG4UpPancreatic juiceNDRG4 promoterMethylationD
SSTUpPlasmaSSTDownstream signaling, transcriptional regulationP
TFPI2UpPancreatic juiceTFPI2Gene silencing, cell adhesion plasmin signalingP
TAC1UpPlasmaTAC1Gpcr downstream, signal transductionP
VIMUpPlasmaVimentinCell signalingP
WNT5AUpPlasmaWNT5AWnt signalingP
PENKUpPancreatic juiceProenkephalinApoptotic, downstream signaling
DCCDownPlasmaNetrin-1 ReceptorMutationD
P16DownPancreatic juiceCDKN2AMutation, cell cycle regulationD
P14DownPlasmaCDKN2ACell cycleD
DNMT1UpPlasmaDNMT1MutationD
DNMT3AUpPlasmaDNMT3AMethylationD
DNMT3BUpPlasmaDNAMethylationD

Proteomic techniques are gaining a lot of attention for studying protein expression in PanCa. Urine as a source for the non-invasive analysis of biomarkers has gained attention. Advantages of using urine are: (1) Common proteins such as albumin are less abundant; (2) Sample processing is simpler (the plasma secreted proteome (secretome) is much more complex than the urine proteome); (3) Urine is more thermostable; and (4) 49% of proteins are soluble products in urine obtained from glomerular filtration of lymphatic fluids (indicates that various external sources contribute towards the pool of the urine proteome and its protein components). Similar studies have been completed in breast and ovarian cancers; and it is likely to remain an active area of investigation (Table 2)[47,68,69].

Role of secretome as a diagnostic and therapeutic biomarker

Cell free nucleic acid alone or combined with the secretome is likely to enhance sensitivity in PanCa diagnosis, and it remains an area of high interest in the field. Research often claims that patients with some signature of circulating proteins ideally overlap with circulating DNA. The main issue is that cfDNA and cell free proteins are most often seen in advanced stages than in early stages[63]. A secretome study to identify cell free proteins as biomarkers in PanCa is usually dominated by the urine proteome. In a study by Poruk et al[70], the researchers validated a pair of biomarkers to distinguish between PDAC and healthy controls as well as distinguishing it from CP. This group also focused on the ability of the biomarker to identify the early onset of the disease. The pair of biomarkers were OPN and TIMP-1, and they were found to be uniformly overexpressed in the case of PDAC. Due to their tendency to be available as the secretome in blood, they were easy to detect and were shown to be a useful tool for diagnosis. OPN particularly acts as a more advanced prognostic biomarker by itself[70-72]. Urine samples are also a reliable source of cell free biomarker isolation and identification.

A three-protein biomarker panel was developed from urine samples of PDAC patients and were assayed using GeLC/mass spectroscopy (MS)/MS and validated using an enzyme-linked immunosorbent assay. The candidate protein biomarkers were namely LYVE-1, TFF1 and REG1A, and they provided an AUC of 0.89. These biomarkers were able to distinguish between early-stage PDAC and advanced-stage PDAC and were almost consistent with CA-19 levels that were documented in regard to every PDAC stage. The panel was also able to distinguish between PDAC and CP based on the higher levels of biomarkers in PDAC than in CP as well as PDAC stage I-II. The concentrations of these proteins were significantly higher than CP, which generated a clear demarcation between these groups[73].

Kaur et al[74] reported MUC5AC as a suitable biomarker candidate for PanCa diagnosis based on its capability to differentiate between PanCa, CP and AIP. Another urine-based diagnosis study claimed that neutrophil gelatinase-associated lipocalin can provide valuable information on the onset of PanCa identification[21,74]. Considering that pancreatic juice collection is an invasive process, the parallel study was completed studying the role of ARG2 as PanCa biomarker, which was specific in targeting the bile component for PanCa early-stage detection. sLR11, which is present in the bile of PanCa cases, was able to discriminate between PanCa and healthy controls[75]. A panel of CA19-9, CA 242, CA 125 and CEA obtained from serum showed a sensitivity and specificity of 90% and 94%, respectively[76,77]. A novel nucleosome marker panel developed by Bauden et al[36] showed better sensitivity than CA19-9 in discriminating PanCa from a healthy control. The markers were H3K4Me2, H2AZ, H2AK119Ub, H2A1.1 and 5mC[36]. Additionally, as documented by Melo et al[78], the absolute prediction (AUC = 1.0) of PanCa using glypican 1 on exosomes identified by MS showed utility as a biomarker (Table 2).

cfRNA in PanCa and its association with benign pancreatic diseases

Amidst the traditional serum-based biomarkers like CA19-9, CA125 and CEA for PanCa diagnostics, there is an immense need for the development of more sensitive and specific non-invasive biomarkers. Sensitivity and specificity are usually higher when the three are used jointly (Table 3)[21,31].

Table 3 Cell-free RNA involved in pancreatic cancer diagnostic and prognostics.
Target candidate
Up/down
Sample
Target
Pathway
Potential value
ERBB2UpPlasmaCh 12, KRASMutationD
CDKN2ADownPlasmaGene, Ch 9p21MutationD
SMAD4DownPlasmaTGFβCell signalingD
TP53DownPlasmaP53 proteinMutation/cell signalingD
EGFRUpPlasmaExon 19, 21Cell signaling/mutationD
PBRM1DownPlasmaSWI/SNF ch remodeling compMutation, immune checkpoint blockadeD
KMT2DDownPlasmaFOX1-miR-1224, HIF/GATA5-miR-133aMutationD
RNF43DownPlasmaRNF43PROTEINWnt signalingD
TP63DownPlasmaP63 proteinCell signalingD
MTORUpPlasmamTORmTOR signalingD
NRASUpPlasmaNRASMutationD
HRASUpPlasmaHRASMutationD
BRCA1DownPlasmaBRCA1SignalingD
BRCA2DownPlasmaBRCA2SignalingD
PALB2UpPlasmaPALB2Signaling/mutationD
GPC1UpSerumMAPK, PI3K-AKT-mTOR, Hedgehog signalingCell signalingD
PFAAUpSerumMAPK, PI3K-AKT-mTOR, Hedgehog signalingCell signalingD
OPNT+TIMP-1UpSerumMAPK, PI3K-AKT-mTOR, signalingCell signalingD
CPA4UpSerumMAPK, mTOR, Hedgehog signalingCell signalingD
MUC5 ACUpUrine/pancreatic juiceMAPK, PI3K-AKT-mTOR, Hedgehog signalingCell signalingD/P
C4BPAUpSerumMAPK, PI3K-AKT-mTOR, Hedgehog signalingCell signalingD
OPNUpSerumOPNCell signalingP
TIMP-1UpSerumTIMP-1Cell signalingD
NGALUpUrineReceptor binding proteinCell signalingD
ARG2DownPancreatic juiceBeta cell functionCell signalingD
LYVE-1UpUrineReceptor binding glycoproteinD
TFF1UpUrineRegulation of transcription factorPromoter modelingD
REG1AUpUrineREG1A proteinCell signalingD

Uncontrolled proliferating cancer has a high mortality rate, and treatment and diagnosis are also difficult due to the lack of screening imaging modalities and specific biomarkers. Emerging evidence of biomarkers generated from tumor cells contains a high quantity of RNA in the bloodstream that block RNases in blood. They also showcase sufficient levels for quantitative analyses[79]. Living cells consistently release RNA, which is encapsulated inside big lipoprotein complexes, namely exosomes/microvesicles (MVs). RNA that is obtained from dead or dying cells found from blood is connected with apoptotic bodies and protein complexes[80].

Exosomes range from 40-140 nm in diameter and are made of lipoprotein membranous vesicles of endocytic origin. They are formed from the fusion of multivesicular bodies with the plasma membrane. They are released into extracellular spaces thereafter[81]. MVs are much larger than exosomes and have a diameter ranging from 100-1000 nm. They have heterogeneous morphologies, and they originate from the plasma membrane by budding with the extracellular space.

Apoptotic bodies are morphologically MVs that are heterogeneous in shape, spanning 50-500 nm in diameter, and contain organelles. They are released via outward protrusion from the plasma membrane during the late phase of apoptosis[82]. Several studies have demonstrated that tumors specifically secrete exosomes or MVs, which contain specific miRNAs. However, Arroyo et al[83] stated that miRNAs in circulation are principally found in the Ago2 ribonucleoprotein complex and not vesicles. This indicates that detected miRNAs are derived mostly from apoptotic and necrotic processes, which occur frequently in tumor cells. These circulating RNAs are found in the plasma or serum of patients and are upregulated when compared to healthy patients.

A significant amount of RNA biomarkers have been shown to be circulating miRNAs and are mostly tissue specific. Some reports found tumor-specific upregulation of various types of small noncoding RNAs (ncRNAs), such as small nuclear RNAs, Piwi-interacting RNAs and long ncRNAs (lncRNAs) like HOTAIR and MALAT1. They act as diagnostic and prognostic markers in the serum or plasma of cancer patients[84,85]. Amidst that, some have reported alterations in gene expression according to disease progression, which discriminates between a chronic inflammatory state and carcinoma (Table 3)[86,87].

Funaki et al[88] detected CEA mRNA by real-time reverse transcription PCR in the whole blood of PanCa patients. Clarke et al[89] also detected EGFR mRNA in the serum and detected alpha 1,4-N-acetylglucosaminyltransferase mRNA in the mononuclear cell fraction of peripheral blood from PanCa patients. Kang et al[90] further demonstrated that serum-type collagen mRNA acts as good marker of PanCa. It undergoes tumor-specific alternative splicing, which is expected to result in high specificity (Table 3).

In PanCa, the tumor suppressor genes p53, p16 and SMAD4 are rendered inactive by the miRNAs, which are produced only in tumor tissues[91]. In pancreatic cell lines and tissue, miR-21 is increased, which drastically lowers the survival rate. In the KRAS (G12D) animal model, it has been reported that miR-21 overexpression is the lesion onset point that leads to tumor growth. miR-155 is also overexpressed in PanCa. This aids in the development of tumors. Decreasing the expression of miR-155, MT1-MMP, KRAS, and EGFR inhibits cell growth. miR-221 is also upregulated in PanCa and promotes distant metastasis as well as unresectable tumors[92]. In approximately 50%-70% of cases of PanCa in humans, the p53 gene has a point mutation. Hypoxia with malnutrition trigger stress signaling in cells that upregulates p53, which increases the expression of numerous genes, including MIR-107, MIR-34a/b/c and MIR-34 (Table 3)[15,65,86].

Role of cfRNA as a diagnostic and therapeutic biomarker

According to some reports, miRNA dysregulation has been reported in feces, urine and saliva, which are easy to obtain by non-invasive methods. The levels of miR-143, miR-223 and miR-30 were seen to be higher in the urine of patients with stage I disease. The combination of miR-143 with miR-30 showed very high sensitivity and specificity of 83.3% and 96.2%, respectively. Yang et al[93] reported that the levels of miR-21 and miR-155 were much higher in PanCa patient stool when compared with healthy controls. Recently studies revealed that salivary miRNAs were stable because of the protection of exosomes or protein complexes, thus highlighting promising roles as diagnostic markers. SNHG15 serum expression exerts an average diagnostic value (Table 3)[94].

Liao et al[95] found that plasma snoRNA SNORD33/66/76 might serve as a diagnostic biomarker for non-small cell lung cancer (NSCLC) using comprehensive next generation sequencing analysis. Wang et al[96] detected the expression of miRNAs in the plasma of patients with PDAC and identified miRs-21, miR-210, miR-155 and miR-196a. They are upregulated in PanCa tissue and cell lines as candidate biomarkers, and miR-200a/b, miR-18a, miR-221 and miR-196a/b have been found to be upregulated in the serum/plasma in parallel with cancer tissues by comprehensive sequence and microarray analysis (Table 3)[96,97].

Kishikawa et al[86] suggested that some miRNAs, such as miR-16, miR-223 and let-7, are highly and constantly expressed in the serum/plasma and are correlated with the number of blood cells. Tang et al[98] found higher levels of three lncRNAs, long intergenic non protein-coding RNA 1627 (LINC01627), LINC01628 and ERICH1 antisense RNA 1 (ERICH1-AS1; also known as DLGAP2), in the plasma from patients with NSCLC than in samples from healthy individuals after microarray assay. They proposed a potential diagnostic signature for NSCLC based on these lncRNAs. Anfossi et al[99,100] suggested that urine levels of miR-148a and miR-375 have been found to be significantly downregulated and upregulated, respectively, in patients with prostate cancer and enabled patients with prostate cancer to be distinguished from healthy individuals (Table 3).

Higher levels of expression of hsa-miR-106a, hsa-miR-125a-5p, hsa-miR-129-3p, hsa-miR-375, hsa-miR-205, hsa-miR-29b, hsa-miR-21 and hsa-miR-7 in histopathology as well as in cytology samples using real-time reverse transcription PCR act as valuable diagnostic biomarkers in different types of cancer[101]. Also, miR-21, miR-210, miR-155 and miR-196a, which have been upregulated in PanCa tissue and cell lines, have been identified by Wang et al[96] as candidate biomarkers. A possible diagnostic biomarker for malignant melanoma is miR-221. Li et al[103] found that patients with high serum miR-221 levels had significantly worse 5-year relapse-free survival (12.5% vs 45.2%; P = 0.008). Increased circulating levels of miR-221 allowed patients with melanoma to be discriminated from healthy individuals (P = 0.0001) (Table 3)[103].

Rac1 downregulation leads to the switching of the MKK4-JNK-c-Jun pathway, which silences miR-124. In response to TGF, miR-200a and miR-205 are downregulated in PDAC during epithelial-mesenchymal transformation (EMT), while miR-21 overexpression is identified as the lesion initiator that drives tumor growth in a KRAS (G12D) animal model. miR-155, which is elevated in PanCa, aids in the development of tumors.

Karandish et al[104] suggested that stress signaling in cells induced by hypoxia and starvation upregulate p53 and activate the expression of several genes, such as MIR-107, MIR-34a/b/c and MIR-34. Mutation of p53 mediates transcription of MIR-130b and MIR-155, which modifies the expression of the corresponding target genes (ZEB1 and ZNF652). This leads to cell proliferation and invasion in several cancers. ZEB1 has been shown by Wellner et al[106] to control the activation of EMT in pancreatic cancer stem cells and to suppress the production of the miR-200 family[104]. In addition, p53 mutation impairs the maturation of miR-145 and miR-16-1 causing cell proliferation, invasion and migration in PDAC (Table 3).

As a long intergenic non-protein coding RNA, p53 induced transcript (Linc-pint) is ubiquitously expressed in humans and acts as a direct p53 transcriptional target. Plasma lncRNA Linc-pint was significantly decreased in patients with PanCa, carcinoma of the ampulla of Vater and cholangiocarcinoma compared with healthy volunteers[108]. Therefore, Linc-pint might be used for identifying the cause of malignant obstructive jaundice and helping to trace the origin of the cancer. Some of these results indicate that low plasma Linc-pint expression could serve as a minimally invasive biomarker for early PanCa detection and that low Linc-pint levels in PanCa tumors could be used for predicting patient prognosis(Table 3)[103].

Prior studies revealed that upregulation of chromatin-interacting lncRNA along with MEG3 inhibited PanCa cell proliferation with the activation of p53. Additionally, lncRNA-NUTF2P3-001 contributed to PanCa proliferation and invasion by activating the miR-3923/KRAS signaling pathway. Moreover, LOC389641 promoted the progression of PDAC and increased cell invasion by regulating E-cadherin with the possible involvement of TNFRSF10A (Table 3)[109].

The d-signature comprises eight extracellular vesicle long RNAs, including FGA, KRT19, HIST1H2BK, ITIH2, MARCH2, CLDN1, MAL2 and TIMP1, for PDAC detection, and the d-signature could distinguish between CA19-9 negative PDAC cases and healthy controls, thus complementing the use of CA19-9 in PDAC detection. It could also distinguish PDAC from CP with high accuracy. Circ-LDLRAD3 was found to be increased in PanCa tissues as well as in plasma samples. It was markedly related to lymphatic invasion, along with venous invasion and metastasis. Circ-LDLRAD3 has not been reported as an ideal standalone biomarker but may be in combination with CA19-9[110].

The circRNAs PDE8A and IARS are present in the plasma exosome. They were upregulated along with a concurrent association with prognosis and progression of PanCa. They were also likely to be promising biomarkers in the detection of early-stage PanCa. When compared with the healthy controls, circ-001569 levels were higher. Aberrant levels of CPA4, PFAA, MUC5AC, GPANCA1, OPNT + TIMP-1 and C4BPA were frequently reported in serum samples of PanCa patients. In pancreatic juice samples, the higher regulation of ARG2 signifies its role as a biomarker for PanCa diagnosis. Neutrophil gelatinase-associated lipocalin in urine also provides some clue for early diagnosis of PanCa (Table 3)[111,112].

Role of immunological treatments in PanCa and non-malignant diseases

The lack of immune and targeted molecular therapies is a reason for treatment failure and lower survival rates[113]. The reason behind less efficient immune therapies in PanCa is unclear. Usually, PTs are surrounded by thick desmoplastic stroma making targeted treatments less effective. Other reasons of immune therapy failure are chemokine-initiated barring of T cells followed by comparatively faulty antigenicity when compared to other solid tumors[114]. A model states that the systemic progression of PDAC development initiates from PanIN1 to PanIN3 and finally leads to invasive PDAC. This occurs when high progressive deposition of desmoplastic stroma and alteration of leucocyte infiltration happens[115].

The lower mutation burden in PDAC is partially due to poor antigenicity of the tumor microenvironment (TME). The abundant immunosuppressive TME plays a crucial role in blocking T cell activation, which further leads to lower immune therapy efficiency[116]. A significantly increased level of the TH17 and Th1 genes acts as an inflammatory immune marker[117]. A variety of immune biomarkers have been reported in PDAC. Those with sound prognostic capability are CD3, CD4, CD8 and CD20. In contrast, increased expression of CD163, FOXP3, CD204, CD68 and CD66b leads to poorer outcomes[114,118].

Interestingly, the presence of T cells has no clinical correlation with PDAC. Tumor-infiltrating CD3+ T cells have been identified in isolated PDAC tumors. However, due to the lack of CD3 zeta expression, the capability to activate T cell-associated receptor signaling has been lost and is under functioning[119]. The tumor immune microenvironment and TME of PDAC is peculiar when compared with other solid tumors because immune therapy does not yield significant results in PanCa treatment. PD-L1, CTLA-4 and PD-1 targeting antibodies are part of immune checkpoints, and inhibition of these targets has been successful in other malignancies but have failed in curative treatment of PanCa[120]. Of note, PD-L1 and PD-1 expression is significantly lower in PDAC than in other tumors. PD-L1 has high expression in almost 30%-40% of PanCa cases, which is correlated with CD8+ cells, thereby indicating a poor prognosis[120,121].

CONCLUSION

Currently, a variety of specimens have emerged as possible sources for liquid biopsy analysis for diagnostic applications. Apart from plasma or serum, urine and sputum have also produced promising disease diagnostic results[44,122]. Liquid biopsies are ideally non-invasive or minimally invasive for sample collection, minimizing patient stress and discomfort. Non-invasive approaches can significantly aid clinical research and utility. Even the recurrence and metastasis of the disease can be potentially monitored based on the cfDNA methods (Table 4).

Table 4 Cell-free biomarkers involved in distinguishing pancreatic cancer from other pancreatic benign diseases.
Biomarkers
CP
AIP
PanIN
IPMN
MCN
PENK DNAYesNoYesYesYes
CUX2YesNoNoNoNo
REGIAYesNoNoNoNo
TFP12YesNoYesYesYes
FOXE1YesNoYesYesYes
P16YesNoYesYesYes
NPTX2YesNoYesYesYes
PENKYesNoYesYesYes
CYCLIN D2YesNoYesYesYes
CDO1NoYesNoNoNo
HAS-mIR-23aNoNoNoYesNo
HAS-mIR-23bNoNoNoYesNo
HAS-mIR-210YesNoNoNoNo
Let-7cYesNoNoNoNo
S100A9YesNoNoNoNo
BNCINoNoYesNoNo
ADAMTS1NoNoNoNoNo
BNC1NoNoNoNoNo
9-SepNoNoNoNoNo
GPC1NoNoNoNoNo
PFAANoNoNoNoNo
OPNT + TIMP-1NoNoNoNoNo
CPA4NoNoNoNoNo
MUC5 ACNoNoNoNoNo
C4BPANoNoNoNoNo

Therefore, in this review we highlighted the current development in tools for PanCa early diagnosis and prognosis. We demonstrated that useful data can be obtained based on DNA methylation signatures and mutations. The heterogeneity in the pool of cfDNA isolated from a suitable source, such as plasma, contains cfDNA molecules obtained from various origins, and various outcomes, such as DNA methylation or gene mutations, could be obtained from such material. The process of properly understanding the disease source and cfDNA origin has been facilitated significantly by advancements in technologies, such as single cell-based techniques[40]. These advancements can further facilitate liquid biopsy research associated with PanCa biomarker discovery. This also showcases that aberrant DNA methylation signatures harness a significant capability to guide PanCa disease management[32,51]. Approximately 300 clinical trials are currently ongoing that target the use of cfDNA biomarkers in the diagnosis of malignancies. Limitations include cfDNA isolation and sample processing and understanding the biology of ctDNA based on its release from tumors.

The outcome of the secretome can also support PanCa diagnosis significantly. Premalignant lesions such as PanIN are still facing detection challenges because no PanIN-specific protein has been clinically approved. The alterations in cell free nucleic acids does not always manifest into protein changes that can be clinically validated and utilized. However, the secretome in PanCa and secretome biomarkers specific to premalignant lesions are yet to be identified for clinical use but are likely to be highly useful in the future. Validation of biomarkers based on cfDNA alterations and subsequent protein expression can substantially help in the early diagnosis of PanCa and other pancreatic-associated diseases[51,123].

ACKNOWLEDGEMENTS

First, we would like to thank all the authors and clinicians whose valuable work has supported us in writing this review. Second, we also acknowledge clinicians from Tata Medical Center namely Dr. Sudeep Banerjee and Dr. Paromita Roy along with clinicians from Apollo Multispecialty Hospital namely Dr. Sumit Gulati and Dr. Supriyo Ghatak. Dr. Aniruddha Chatterjee would like to thank Rutherford Discovery Fellowship from the Royal Society of New Zealand and University of Otago, Dunedin, New Zealand for their support. The authors specifically thank and acknowledge Mr. Saraswan Sikdar.

Footnotes

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

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: India

Peer-review report’s scientific quality classification

Grade A (Excellent): A

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Białek Ł, Poland; Wang XB, China S-Editor: Zhang H L-Editor: Filipodia P-Editor: Zhang H

References
1.  Sharma S, Tapper WJ, Collins A, Hamady ZZR. Predicting Pancreatic Cancer in the UK Biobank Cohort Using Polygenic Risk Scores and Diabetes Mellitus. Gastroenterology. 2022;162:1665-1674.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 18]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
2.  Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4235]  [Cited by in F6Publishing: 8748]  [Article Influence: 4374.0]  [Reference Citation Analysis (2)]
3.  Kavaliauskas P, Dulskas A, Kildusiene I, Arlauskas R, Stukas R, Smailyte G. Trends in Pancreatic Cancer Incidence and Mortality in Lithuania, 1998-2015. Int J Environ Res Public Health. 2022;19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
4.  Sikdar N, Saha G, Dutta A, Ghosh S, Shrikhande SV, Banerjee S. Genetic Alterations of Periampullary and Pancreatic Ductal Adenocarcinoma: An Overview. Curr Genomics. 2018;19:444-463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
5.  Saha G, Singh R, Mandal A, Das S, Chattopadhyay E, Panja P, Roy P, DeSarkar N, Gulati S, Ghatak S, Ghosh S, Banerjee S, Roy B, Chaudhuri D, Arora N, Biswas NK, Sikdar N. A novel hotspot and rare somatic mutation p.A138V, at TP53 is associated with poor survival of pancreatic ductal and periampullary adenocarcinoma patients. Mol Med. 2020;26:59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
6.  Vasantharajan SS, Eccles MR, Rodger EJ, Pattison S, McCall JL, Gray ES, Calapre L, Chatterjee A. The Epigenetic landscape of Circulating tumour cells. Biochim Biophys Acta Rev Cancer. 2021;1875:188514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
7.  Kilgour E, Rothwell DG, Brady G, Dive C. Liquid Biopsy-Based Biomarkers of Treatment Response and Resistance. Cancer Cell. 2020;37:485-495.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 200]  [Article Influence: 50.0]  [Reference Citation Analysis (0)]
8.  Krebs MG, Metcalf RL, Carter L, Brady G, Blackhall FH, Dive C. Molecular analysis of circulating tumour cells-biology and biomarkers. Nat Rev Clin Oncol. 2014;11:129-144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 483]  [Cited by in F6Publishing: 452]  [Article Influence: 45.2]  [Reference Citation Analysis (0)]
9.  Chen D, Xu T, Wang S, Chang H, Yu T, Zhu Y, Chen J. Liquid Biopsy Applications in the Clinic. Mol Diagn Ther. 2020;24:125-132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 27]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
10.  Qi ZH, Xu HX, Zhang SR, Xu JZ, Li S, Gao HL, Jin W, Wang WQ, Wu CT, Ni QX, Yu XJ, Liu L. The Significance of Liquid Biopsy in Pancreatic Cancer. J Cancer. 2018;9:3417-3426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 36]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
11.  Siravegna G, Mussolin B, Venesio T, Marsoni S, Seoane J, Dive C, Papadopoulos N, Kopetz S, Corcoran RB, Siu LL, Bardelli A. How liquid biopsies can change clinical practice in oncology. Ann Oncol. 2019;30:1580-1590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 211]  [Article Influence: 42.2]  [Reference Citation Analysis (0)]
12.  Liu S, Wang J. Current and Future Perspectives of Cell-Free DNA in Liquid Biopsy. Curr Issues Mol Biol. 2022;44:2695-2709.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 11]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
13.  Yokode M, Shiokawa M, Kodama Y. Review of Diagnostic Biomarkers in Autoimmune Pancreatitis: Where Are We Now? Diagnostics (Basel). 2021;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
14.  Henriksen SD, Thorlacius-Ussing O. Cell-Free DNA Methylation as Blood-Based Biomarkers for Pancreatic Adenocarcinoma-A Literature Update. Epigenomes. 2021;5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
15.  Minaga K, Watanabe T, Hara A, Kamata K, Omoto S, Nakai A, Otsuka Y, Sekai I, Yoshikawa T, Yamao K, Takenaka M, Chiba Y, Kudo M. Identification of serum IFN-α and IL-33 as novel biomarkers for type 1 autoimmune pancreatitis and IgG4-related disease. Sci Rep. 2020;10:14879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
16.  Kawa S, Kamisawa T, Notohara K, Fujinaga Y, Inoue D, Koyama T, Okazaki K. Japanese Clinical Diagnostic Criteria for Autoimmune Pancreatitis, 2018: Revision of Japanese Clinical Diagnostic Criteria for Autoimmune Pancreatitis, 2011. Pancreas. 2020;49:e13-e14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 65]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
17.  Kamisawa T, Chari ST, Lerch MM, Kim MH, Gress TM, Shimosegawa T. Recent advances in autoimmune pancreatitis: type 1 and type 2. Gut. 2013;62:1373-1380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 145]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
18.  Chatterjee A, Bararia A, Ganguly D, Mondal PK, Roy P, Banerjee S, Ghosh S, Gulati S, Ghatak S, Chattopadhay BK, Basu P, Chatterjee A, Sikdar N. DNA methylome in pancreatic cancer identified novel promoter hyper-methylation in NPY and FAIM2 genes associated with poor prognosis in Indian patient cohort. Cancer Cell Int. 2022;22:334.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
19.  Bararia A, Dey S, Gulati S, Ghatak S, Ghosh S, Banerjee S, Sikdar N. Differential methylation landscape of pancreatic ductal adenocarcinoma and its precancerous lesions. Hepatobiliary Pancreat Dis Int. 2020;19:205-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
20.  Cohen JD, Li L, Wang Y, Thoburn C, Afsari B, Danilova L, Douville C, Javed AA, Wong F, Mattox A, Hruban RH, Wolfgang CL, Goggins MG, Dal Molin M, Wang TL, Roden R, Klein AP, Ptak J, Dobbyn L, Schaefer J, Silliman N, Popoli M, Vogelstein JT, Browne JD, Schoen RE, Brand RE, Tie J, Gibbs P, Wong HL, Mansfield AS, Jen J, Hanash SM, Falconi M, Allen PJ, Zhou S, Bettegowda C, Diaz LA Jr, Tomasetti C, Kinzler KW, Vogelstein B, Lennon AM, Papadopoulos N. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359:926-930.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1752]  [Cited by in F6Publishing: 1590]  [Article Influence: 265.0]  [Reference Citation Analysis (0)]
21.  Wu H, Ou S, Zhang H, Huang R, Yu S, Zhao M, Tai S. Advances in biomarkers and techniques for pancreatic cancer diagnosis. Cancer Cell Int. 2022;22:220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 18]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
22.  Banerjee R, Smith J, Eccles MR, Weeks RJ, Chatterjee A. Epigenetic basis and targeting of cancer metastasis. Trends Cancer. 2022;8:226-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
23.  Smith J, Sen S, Weeks RJ, Eccles MR, Chatterjee A. Promoter DNA Hypermethylation and Paradoxical Gene Activation. Trends Cancer. 2020;6:392-406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 113]  [Article Influence: 28.3]  [Reference Citation Analysis (0)]
24.  Galardi F, Luca F, Romagnoli D, Biagioni C, Moretti E, Biganzoli L, Leo AD, Migliaccio I, Malorni L, Benelli M. Cell-Free DNA-Methylation-Based Methods and Applications in Oncology. Biomolecules. 2020;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
25.  Liu MC, Oxnard GR, Klein EA, Swanton C, Seiden MV; CCGA Consortium. Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann Oncol. 2020;31:745-759.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 707]  [Cited by in F6Publishing: 662]  [Article Influence: 165.5]  [Reference Citation Analysis (0)]
26.  Manoochehri M, Wu Y, Giese NA, Strobel O, Kutschmann S, Haller F, Hoheisel JD, Moskalev EA, Hackert T, Bauer AS. SST gene hypermethylation acts as a pan-cancer marker for pancreatic ductal adenocarcinoma and multiple other tumors: toward its use for blood-based diagnosis. Mol Oncol. 2020;14:1252-1267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
27.  Luo H, Wei W, Ye Z, Zheng J, Xu RH. Liquid Biopsy of Methylation Biomarkers in Cell-Free DNA. Trends Mol Med. 2021;27:482-500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 127]  [Article Influence: 42.3]  [Reference Citation Analysis (0)]
28.  Huang Y, Huang C, Jiang X, Yan Y, Zhuang K, Liu F, Li P, Wen Y. Exploration of Potential Roles of m5C-Related Regulators in Colon Adenocarcinoma Prognosis. Front Genet. 2022;13:816173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
29.  Luo H, Zhao Q, Wei W, Zheng L, Yi S, Li G, Wang W, Sheng H, Pu H, Mo H, Zuo Z, Liu Z, Li C, Xie C, Zeng Z, Li W, Hao X, Liu Y, Cao S, Liu W, Gibson S, Zhang K, Xu G, Xu RH. Circulating tumor DNA methylation profiles enable early diagnosis, prognosis prediction, and screening for colorectal cancer. Sci Transl Med. 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 221]  [Article Influence: 55.3]  [Reference Citation Analysis (0)]
30.  Tivey A, Church M, Rothwell D, Dive C, Cook N. Circulating tumour DNA - looking beyond the blood. Nat Rev Clin Oncol. 2022;19:600-612.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 92]  [Article Influence: 46.0]  [Reference Citation Analysis (0)]
31.  Goonetilleke KS, Siriwardena AK. Systematic review of carbohydrate antigen (CA 19-9) as a biochemical marker in the diagnosis of pancreatic cancer. Eur J Surg Oncol. 2007;33:266-270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 539]  [Cited by in F6Publishing: 571]  [Article Influence: 31.7]  [Reference Citation Analysis (1)]
32.  Brancaccio M, Natale F, Falco G, Angrisano T. Cell-Free DNA Methylation: The New Frontiers of Pancreatic Cancer Biomarkers' Discovery. Genes (Basel). 2019;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
33.  Guler GD, Ning Y, Ku CJ, Phillips T, McCarthy E, Ellison CK, Bergamaschi A, Collin F, Lloyd P, Scott A, Antoine M, Wang W, Chau K, Ashworth A, Quake SR, Levy S. Detection of early stage pancreatic cancer using 5-hydroxymethylcytosine signatures in circulating cell free DNA. Nat Commun. 2020;11:5270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 76]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
34.  Moss J, Magenheim J, Neiman D, Zemmour H, Loyfer N, Korach A, Samet Y, Maoz M, Druid H, Arner P, Fu KY, Kiss E, Spalding KL, Landesberg G, Zick A, Grinshpun A, Shapiro AMJ, Grompe M, Wittenberg AD, Glaser B, Shemer R, Kaplan T, Dor Y. Comprehensive human cell-type methylation atlas reveals origins of circulating cell-free DNA in health and disease. Nat Commun. 2018;9:5068.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 331]  [Cited by in F6Publishing: 513]  [Article Influence: 85.5]  [Reference Citation Analysis (0)]
35.  Chhatriya B, Mukherjee M, Ray S, Saha B, Lahiri S, Halder S, Ghosh I, Khamrui S, Das K, Bhattacharjee S, Mohapatra SK, Goswami S. Transcriptome analysis identifies putative multi-gene signature distinguishing benign and malignant pancreatic head mass. J Transl Med. 2020;18:420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
36.  Bauden M, Pamart D, Ansari D, Herzog M, Eccleston M, Micallef J, Andersson B, Andersson R. Circulating nucleosomes as epigenetic biomarkers in pancreatic cancer. Clin Epigenetics. 2015;7:106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 72]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
37.  Kisiel JB, Raimondo M, Taylor WR, Yab TC, Mahoney DW, Sun Z, Middha S, Baheti S, Zou H, Smyrk TC, Boardman LA, Petersen GM, Ahlquist DA. New DNA Methylation Markers for Pancreatic Cancer: Discovery, Tissue Validation, and Pilot Testing in Pancreatic Juice. Clin Cancer Res. 2015;21:4473-4481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 91]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
38.  Natale F, Vivo M, Falco G, Angrisano T. Deciphering DNA methylation signatures of pancreatic cancer and pancreatitis. Clin Epigenetics. 2019;11:132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 34]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
39.  Liggett T, Melnikov A, Yi QL, Replogle C, Brand R, Kaul K, Talamonti M, Abrams RA, Levenson V. Differential methylation of cell-free circulating DNA among patients with pancreatic cancer versus chronic pancreatitis. Cancer. 2010;116:1674-1680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 102]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
40.  Lehmann-Werman R, Neiman D, Zemmour H, Moss J, Magenheim J, Vaknin-Dembinsky A, Rubertsson S, Nellgård B, Blennow K, Zetterberg H, Spalding K, Haller MJ, Wasserfall CH, Schatz DA, Greenbaum CJ, Dorrell C, Grompe M, Zick A, Hubert A, Maoz M, Fendrich V, Bartsch DK, Golan T, Ben Sasson SA, Zamir G, Razin A, Cedar H, Shapiro AM, Glaser B, Shemer R, Dor Y. Identification of tissue-specific cell death using methylation patterns of circulating DNA. Proc Natl Acad Sci U S A. 2016;113:E1826-E1834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 381]  [Cited by in F6Publishing: 398]  [Article Influence: 49.8]  [Reference Citation Analysis (0)]
41.  Chen R, Pan S, Yi EC, Donohoe S, Bronner MP, Potter JD, Goodlett DR, Aebersold R, Brentnall TA. Quantitative proteomic profiling of pancreatic cancer juice. Proteomics. 2006;6:3871-3879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 98]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
42.  Matsubayashi H, Canto M, Sato N, Klein A, Abe T, Yamashita K, Yeo CJ, Kalloo A, Hruban R, Goggins M. DNA methylation alterations in the pancreatic juice of patients with suspected pancreatic disease. Cancer Res. 2006;66:1208-1217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 170]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
43.  Nishizawa N, Harada H, Kumamoto Y, Kaizu T, Katoh H, Tajima H, Ushiku H, Yokoi K, Igarashi K, Fujiyama Y, Okuwaki K, Iwai T, Watanabe M, Yamashita K. Diagnostic potential of hypermethylation of the cysteine dioxygenase 1 gene (CDO1) promoter DNA in pancreatic cancer. Cancer Sci. 2019;110:2846-2855.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
44.  Humeau M, Vignolle-Vidoni A, Sicard F, Martins F, Bournet B, Buscail L, Torrisani J, Cordelier P. Salivary MicroRNA in Pancreatic Cancer Patients. PLoS One. 2015;10:e0130996.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 91]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
45.  Su Q, Zhu EC, Qu YL, Wang DY, Qu WW, Zhang CG, Wu T, Gao ZH. Serum level of co-expressed hub miRNAs as diagnostic and prognostic biomarkers for pancreatic ductal adenocarcinoma. J Cancer. 2018;9:3991-3999.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 28]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
46.  Martínez-Bosch N, Cristóbal H, Iglesias M, Gironella M, Barranco L, Visa L, Calafato D, Jiménez-Parrado S, Earl J, Carrato A, Manero-Rupérez N, Moreno M, Morales A, Guerra C, Navarro P, García de Frutos P. Soluble AXL is a novel blood marker for early detection of pancreatic ductal adenocarcinoma and differential diagnosis from chronic pancreatitis. EBioMedicine. 2022;75:103797.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 20]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
47.  Weeks ME, Hariharan D, Petronijevic L, Radon TP, Whiteman HJ, Kocher HM, Timms JF, Lemoine NR, Crnogorac-Jurcevic T. Analysis of the urine proteome in patients with pancreatic ductal adenocarcinoma. Proteomics Clin Appl. 2008;2:1047-1057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 32]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
48.  Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, Bartlett BR, Wang H, Luber B, Alani RM, Antonarakis ES, Azad NS, Bardelli A, Brem H, Cameron JL, Lee CC, Fecher LA, Gallia GL, Gibbs P, Le D, Giuntoli RL, Goggins M, Hogarty MD, Holdhoff M, Hong SM, Jiao Y, Juhl HH, Kim JJ, Siravegna G, Laheru DA, Lauricella C, Lim M, Lipson EJ, Marie SK, Netto GJ, Oliner KS, Olivi A, Olsson L, Riggins GJ, Sartore-Bianchi A, Schmidt K, Shih lM, Oba-Shinjo SM, Siena S, Theodorescu D, Tie J, Harkins TT, Veronese S, Wang TL, Weingart JD, Wolfgang CL, Wood LD, Xing D, Hruban RH, Wu J, Allen PJ, Schmidt CM, Choti MA, Velculescu VE, Kinzler KW, Vogelstein B, Papadopoulos N, Diaz LA Jr. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6:224ra24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2770]  [Cited by in F6Publishing: 3188]  [Article Influence: 318.8]  [Reference Citation Analysis (0)]
49.  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: ]
50.  Shapiro B, Chakrabarty M, Cohn EM, Leon SA. Determination of circulating DNA levels in patients with benign or malignant gastrointestinal disease. Cancer. 1983;51:2116-2120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
51.  Chang S, Hur JY, Choi YL, Lee CH, Kim WS. Current status and future perspectives of liquid biopsy in non-small cell lung cancer. J Pathol Transl Med. 2020;54:204-212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
52.  Thakral D, Das N, Basnal A, Gupta R. Cell-free DNA for genomic profiling and minimal residual disease monitoring in Myeloma- are we there yet? Am J Blood Res. 2020;10:26-45.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Chatterjee A, Stockwell PA, Ahn A, Rodger EJ, Leichter AL, Eccles MR. Genome-wide methylation sequencing of paired primary and metastatic cell lines identifies common DNA methylation changes and a role for EBF3 as a candidate epigenetic driver of melanoma metastasis. Oncotarget. 2017;8:6085-6101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 47]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
54.  Chatterjee A, Rodger EJ, Eccles MR. Epigenetic drivers of tumourigenesis and cancer metastasis. Semin Cancer Biol. 2018;51:149-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 198]  [Article Influence: 28.3]  [Reference Citation Analysis (0)]
55.  Melnikov AA, Scholtens D, Talamonti MS, Bentrem DJ, Levenson VV. Methylation profile of circulating plasma DNA in patients with pancreatic cancer. J Surg Oncol. 2009;99:119-122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 62]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
56.  Henriksen SD, Madsen PH, Larsen AC, Johansen MB, Pedersen IS, Krarup H, Thorlacius-Ussing O. Promoter hypermethylation in plasma-derived cell-free DNA as a prognostic marker for pancreatic adenocarcinoma staging. Int J Cancer. 2017;141:2489-2497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
57.  Sato N, Fukushima N, Maitra A, Matsubayashi H, Yeo CJ, Cameron JL, Hruban RH, Goggins M. Discovery of novel targets for aberrant methylation in pancreatic carcinoma using high-throughput microarrays. Cancer Res. 2003;63:3735-3742.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Yi JM, Guzzetta AA, Bailey VJ, Downing SR, Van Neste L, Chiappinelli KB, Keeley BP, Stark A, Herrera A, Wolfgang C, Pappou EP, Iacobuzio-Donahue CA, Goggins MG, Herman JG, Wang TH, Baylin SB, Ahuja N. Novel methylation biomarker panel for the early detection of pancreatic cancer. Clin Cancer Res. 2013;19:6544-6555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 111]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
59.  Li S, Yuan Y, Xiao H, Dai J, Ye Y, Zhang Q, Zhang Z, Jiang Y, Luo J, Hu J, Chen C, Wang G. Discovery and validation of DNA methylation markers for overall survival prognosis in patients with thymic epithelial tumors. Clin Epigenetics. 2019;11:38.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 17]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
60.  Li XB, Ma J, Liu ZW, He WF, Li ZZ, Cui M, Hao H, Zhou GP, You L, Wang J, Han XL, Zhao YP. Non-invasive detection of pancreatic cancer by measuring DNA methylation of Basonuclin 1 and Septin 9 in plasma. Chin Med J (Engl). 2019;132:1504-1506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
61.  Rodger EJ, Chatterjee A, Morison IM. 5-hydroxymethylcytosine: a potential therapeutic target in cancer. Epigenomics. 2014;6:503-514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
62.  Cao F, Wei A, Hu X, He Y, Zhang J, Xia L, Tu K, Yuan J, Guo Z, Liu H, Xie D, Li A. Integrated epigenetic biomarkers in circulating cell-free DNA as a robust classifier for pancreatic cancer. Clin Epigenetics. 2020;12:112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
63.  Cohen JD, Javed AA, Thoburn C, Wong F, Tie J, Gibbs P, Schmidt CM, Yip-Schneider MT, Allen PJ, Schattner M, Brand RE, Singhi AD, Petersen GM, Hong SM, Kim SC, Falconi M, Doglioni C, Weiss MJ, Ahuja N, He J, Makary MA, Maitra A, Hanash SM, Dal Molin M, Wang Y, Li L, Ptak J, Dobbyn L, Schaefer J, Silliman N, Popoli M, Goggins MG, Hruban RH, Wolfgang CL, Klein AP, Tomasetti C, Papadopoulos N, Kinzler KW, Vogelstein B, Lennon AM. Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers. Proc Natl Acad Sci U S A. 2017;114:10202-10207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 386]  [Cited by in F6Publishing: 373]  [Article Influence: 53.3]  [Reference Citation Analysis (0)]
64.  Kim JE, Lee KT, Lee JK, Paik SW, Rhee JC, Choi KW. Clinical usefulness of carbohydrate antigen 19-9 as a screening test for pancreatic cancer in an asymptomatic population. J Gastroenterol Hepatol. 2004;19:182-186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 236]  [Cited by in F6Publishing: 241]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
65.  Mohan S, Ayub M, Rothwell DG, Gulati S, Kilerci B, Hollebecque A, Sun Leong H, Smith NK, Sahoo S, Descamps T, Zhou C, Hubner RA, McNamara MG, Lamarca A, Valle JW, Dive C, Brady G. Analysis of circulating cell-free DNA identifies KRAS copy number gain and mutation as a novel prognostic marker in Pancreatic cancer. Sci Rep. 2019;9:11610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 34]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
66.  Fukutake N, Ueno M, Hiraoka N, Shimada K, Shiraishi K, Saruki N, Ito T, Yamakado M, Ono N, Imaizumi A, Kikuchi S, Yamamoto H, Katayama K. A Novel Multivariate Index for Pancreatic Cancer Detection Based On the Plasma Free Amino Acid Profile. PLoS One. 2015;10:e0132223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 74]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
67.  Sun L, Burnett J, Guo C, Xie Y, Pan J, Yang Z, Ran Y, Sun D. CPA4 is a promising diagnostic serum biomarker for pancreatic cancer. Am J Cancer Res. 2016;6:91-96.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Barratt J, Topham P. Urine proteomics: the present and future of measuring urinary protein components in disease. CMAJ. 2007;177:361-368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 153]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
69.  Schaub S, Wilkins J, Weiler T, Sangster K, Rush D, Nickerson P. Urine protein profiling with surface-enhanced laser-desorption/ionization time-of-flight mass spectrometry. Kidney Int. 2004;65:323-332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 244]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
70.  Poruk KE, Firpo MA, Scaife CL, Adler DG, Emerson LL, Boucher KM, Mulvihill SJ. Serum osteopontin and tissue inhibitor of metalloproteinase 1 as diagnostic and prognostic biomarkers for pancreatic adenocarcinoma. Pancreas. 2013;42:193-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 79]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
71.  Zhivkova-Galunska M, Adwan H, Eyol E, Kleeff J, Kolb A, Bergmann F, Berger MR. Osteopontin but not osteonectin favors the metastatic growth of pancreatic cancer cell lines. Cancer Biol Ther. 2010;10:54-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
72.  Fahrmann JF, Bantis LE, Capello M, Scelo G, Dennison JB, Patel N, Murage E, Vykoukal J, Kundnani DL, Foretova L, Fabianova E, Holcatova I, Janout V, Feng Z, Yip-Schneider M, Zhang J, Brand R, Taguchi A, Maitra A, Brennan P, Max Schmidt C, Hanash S. A Plasma-Derived Protein-Metabolite Multiplexed Panel for Early-Stage Pancreatic Cancer. J Natl Cancer Inst. 2019;111:372-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 73]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
73.  Radon TP, Massat NJ, Jones R, Alrawashdeh W, Dumartin L, Ennis D, Duffy SW, Kocher HM, Pereira SP, Guarner posthumous L, Murta-Nascimento C, Real FX, Malats N, Neoptolemos J, Costello E, Greenhalf W, Lemoine NR, Crnogorac-Jurcevic T. Identification of a Three-Biomarker Panel in Urine for Early Detection of Pancreatic Adenocarcinoma. Clin Cancer Res. 2015;21:3512-3521.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 125]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
74.  Kaur S, Smith LM, Patel A, Menning M, Watley DC, Malik SS, Krishn SR, Mallya K, Aithal A, Sasson AR, Johansson SL, Jain M, Singh S, Guha S, Are C, Raimondo M, Hollingsworth MA, Brand RE, Batra SK. A Combination of MUC5AC and CA19-9 Improves the Diagnosis of Pancreatic Cancer: A Multicenter Study. Am J Gastroenterol. 2017;112:172-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 96]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
75.  Terai K, Jiang M, Tokuyama W, Murano T, Takada N, Fujimura K, Ebinuma H, Kishimoto T, Hiruta N, Schneider WJ, Bujo H. Levels of soluble LR11/SorLA are highly increased in the bile of patients with biliary tract and pancreatic cancers. Clin Chim Acta. 2016;457:130-136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
76.  Ansari D, Torén W, Zhou Q, Hu D, Andersson R. Proteomic and genomic profiling of pancreatic cancer. Cell Biol Toxicol. 2019;35:333-343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 32]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
77.  Gu YL, Lan C, Pei H, Yang SN, Liu YF, Xiao LL. Applicative Value of Serum CA19-9, CEA, CA125 and CA242 in Diagnosis and Prognosis for Patients with Pancreatic Cancer Treated by Concurrent Chemoradiotherapy. Asian Pac J Cancer Prev. 2015;16:6569-6573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 49]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
78.  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: 1991]  [Article Influence: 221.2]  [Reference Citation Analysis (0)]
79.  Tsui NB, Ng EK, Lo YM. Stability of endogenous and added RNA in blood specimens, serum, and plasma. Clin Chem. 2002;48:1647-1653.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Ma R, Jiang T, Kang X. Circulating microRNAs in cancer: origin, function and application. J Exp Clin Cancer Res. 2012;31:38.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 140]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
81.  Samos J, García-Olmo DC, Picazo MG, Rubio-Vitaller A, García-Olmo D. Circulating nucleic acids in plasma/serum and tumor progression: are apoptotic bodies involved? Ann N Y Acad Sci. 2006;1075:165-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 19]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
82.  Sato-Kuwabara Y, Melo SA, Soares FA, Calin GA. The fusion of two worlds: non-coding RNAs and extracellular vesicles--diagnostic and therapeutic implications (Review). Int J Oncol. 2015;46:17-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 174]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
83.  Arroyo JD, Chevillet JR, Kroh EM, Ruf IK, Pritchard CC, Gibson DF, Mitchell PS, Bennett CF, Pogosova-Agadjanyan EL, Stirewalt DL, Tait JF, Tewari M. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci U S A. 2011;108:5003-5008.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2345]  [Cited by in F6Publishing: 2510]  [Article Influence: 193.1]  [Reference Citation Analysis (0)]
84.  Ji P, Diederichs S, Wang W, Böing S, Metzger R, Schneider PM, Tidow N, Brandt B, Buerger H, Bulk E, Thomas M, Berdel WE, Serve H, Müller-Tidow C. MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene. 2003;22:8031-8041.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1584]  [Cited by in F6Publishing: 1735]  [Article Influence: 82.6]  [Reference Citation Analysis (0)]
85.  Saha B, Chhatriya B, Pramanick S, Goswami S. Bioinformatic Analysis and Integration of Transcriptome and Proteome Results Identify Key Coding and Noncoding Genes Predicting Malignancy in Intraductal Papillary Mucinous Neoplasms of the Pancreas. Biomed Res Int. 2021;2021:1056622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
86.  Kishikawa T, Otsuka M, Ohno M, Yoshikawa T, Takata A, Koike K. Circulating RNAs as new biomarkers for detecting pancreatic cancer. World J Gastroenterol. 2015;21:8527-8540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 110]  [Cited by in F6Publishing: 110]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
87.  Chhatriya B, Mukherjee M, Ray S, Sarkar P, Chatterjee S, Nath D, Das K, Goswami S. Comparison of tumour and serum specific microRNA changes dissecting their role in pancreatic ductal adenocarcinoma: a meta-analysis. BMC Cancer. 2019;19:1175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
88.  Funaki NO, Tanaka J, Kasamatsu T, Ohshio G, Hosotani R, Okino T, Imamura M. Identification of carcinoembryonic antigen mRNA in circulating peripheral blood of pancreatic carcinoma and gastric carcinoma patients. Life Sci. 1996;59:2187-2199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 39]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
89.  Clarke LE, Leitzel K, Smith J, Ali SM, Lipton A. Epidermal growth factor receptor mRNA in peripheral blood of patients with pancreatic, lung, and colon carcinomas detected by RT-PCR. Int J Oncol. 2003;22:425-430.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Kang CY, Wang J, Axell-House D, Soni P, Chu ML, Chipitsyna G, Sarosiek K, Sendecki J, Hyslop T, Al-Zoubi M, Yeo CJ, Arafat HA. Clinical significance of serum COL6A3 in pancreatic ductal adenocarcinoma. J Gastrointest Surg. 2014;18:7-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
91.  Chang TC, Wentzel EA, Kent OA, Ramachandran K, Mullendore M, Lee KH, Feldmann G, Yamakuchi M, Ferlito M, Lowenstein CJ, Arking DE, Beer MA, Maitra A, Mendell JT. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol Cell. 2007;26:745-752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1481]  [Cited by in F6Publishing: 1537]  [Article Influence: 90.4]  [Reference Citation Analysis (0)]
92.  Habbe N, Koorstra JB, Mendell JT, Offerhaus GJ, Ryu JK, Feldmann G, Mullendore ME, Goggins MG, Hong SM, Maitra A. MicroRNA miR-155 is a biomarker of early pancreatic neoplasia. Cancer Biol Ther. 2009;8:340-346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 249]  [Article Influence: 16.6]  [Reference Citation Analysis (0)]
93.  Yang JY, Sun YW, Liu DJ, Zhang JF, Li J, Hua R. MicroRNAs in stool samples as potential screening biomarkers for pancreatic ductal adenocarcinoma cancer. Am J Cancer Res. 2014;4:663-673.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Jiang P, Yin Y, Wu Y, Sun Z. Silencing of long non-coding RNA SNHG15 suppresses proliferation, migration and invasion of pancreatic cancer cells by regulating the microRNA-345-5p/RAB27B axis. Exp Ther Med. 2021;22:1273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
95.  Liao J, Yu L, Mei Y, Guarnera M, Shen J, Li R, Liu Z, Jiang F. Small nucleolar RNA signatures as biomarkers for non-small-cell lung cancer. Mol Cancer. 2010;9:198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 190]  [Cited by in F6Publishing: 201]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
96.  Wang H, Yin F, Yuan F, Men Y, Deng M, Liu Y, Li Q. Pancreatic cancer differential methylation atlas in blood, peri-carcinomatous and diseased tissue. Transl Cancer Res. 2020;9:421-431.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
97.  Liu J, Gao J, Du Y, Li Z, Ren Y, Gu J, Wang X, Gong Y, Wang W, Kong X. Combination of plasma microRNAs with serum CA19-9 for early detection of pancreatic cancer. Int J Cancer. 2012;131:683-691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 210]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
98.  Tang Q, Ni Z, Cheng Z, Xu J, Yu H, Yin P. Three circulating long non-coding RNAs act as biomarkers for predicting NSCLC. Cell Physiol Biochem. 2015;37:1002-1009.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 44]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
99.  Anfossi S, Babayan A, Pantel K, Calin GA. Clinical utility of circulating non-coding RNAs - an update. Nat Rev Clin Oncol. 2018;15:541-563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 308]  [Article Influence: 61.6]  [Reference Citation Analysis (0)]
100.  Anfossi S, Giordano A, Gao H, Cohen EN, Tin S, Wu Q, Garza RJ, Debeb BG, Alvarez RH, Valero V, Hortobagyi GN, Calin GA, Ueno NT, Woodward WA, Reuben JM. High serum miR-19a levels are associated with inflammatory breast cancer and are predictive of favorable clinical outcome in patients with metastatic HER2+ inflammatory breast cancer. PLoS One. 2014;9:e83113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 67]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
101.  Kamińska K, Nalejska E, Kubiak M, Wojtysiak J, Żołna Ł, Kowalewski J, Lewandowska MA. Prognostic and Predictive Epigenetic Biomarkers in Oncology. Mol Diagn Ther. 2019;23:83-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 44]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
102.  Akamatsu M, Makino N, Ikeda Y, Matsuda A, Ito M, Kakizaki Y, Saito Y, Ishizawa T, Kobayashi T, Furukawa T, Ueno Y. Specific MAPK-Associated MicroRNAs in Serum Differentiate Pancreatic Cancer from Autoimmune Pancreatitis. PLoS One. 2016;11:e0158669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 23]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
103.  Li L, Zhang GQ, Chen H, Zhao ZJ, Chen HZ, Liu H, Wang G, Jia YH, Pan SH, Kong R, Wang YW, Sun B. Plasma and tumor levels of Linc-pint are diagnostic and prognostic biomarkers for pancreatic cancer. Oncotarget. 2016;7:71773-71781.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 53]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
104.  Karandish F, Mallik S. Biomarkers and Targeted Therapy in Pancreatic Cancer. Biomark Cancer. 2016;8:27-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 25]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
105.  Jia X, Wang X, Guo X, Ji J, Lou G, Zhao J, Zhou W, Guo M, Zhang M, Li C, Tai S, Yu S. MicroRNA-124: An emerging therapeutic target in cancer. Cancer Med. 2019;8:5638-5650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 35]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
106.  Wellner U, Schubert J, Burk UC, Schmalhofer O, Zhu F, Sonntag A, Waldvogel B, Vannier C, Darling D, zur Hausen A, Brunton VG, Morton J, Sansom O, Schüler J, Stemmler MP, Herzberger C, Hopt U, Keck T, Brabletz S, Brabletz T. The EMT-activator ZEB1 promotes tumorigenicity by repressing stemness-inhibiting microRNAs. Nat Cell Biol. 2009;11:1487-1495.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1262]  [Cited by in F6Publishing: 1339]  [Article Influence: 89.3]  [Reference Citation Analysis (0)]
107.  Wang P, Chen L, Zhang J, Chen H, Fan J, Wang K, Luo J, Chen Z, Meng Z, Liu L. Methylation-mediated silencing of the miR-124 genes facilitates pancreatic cancer progression and metastasis by targeting Rac1. Oncogene. 2014;33:514-524.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 175]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
108.  Marín-Béjar O, Marchese FP, Athie A, Sánchez Y, González J, Segura V, Huang L, Moreno I, Navarro A, Monzó M, García-Foncillas J, Rinn JL, Guo S, Huarte M. Pint lincRNA connects the p53 pathway with epigenetic silencing by the Polycomb repressive complex 2. Genome Biol. 2013;14:R104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 208]  [Article Influence: 23.1]  [Reference Citation Analysis (0)]
109.  Zheng S, Chen H, Wang Y, Gao W, Fu Z, Zhou Q, Jiang Y, Lin Q, Tan L, Ye H, Zhao X, Luo Y, Li G, Ye L, Liu Y, Li W, Li Z, Chen R. Long non-coding RNA LOC389641 promotes progression of pancreatic ductal adenocarcinoma and increases cell invasion by regulating E-cadherin in a TNFRSF10A-related manner. Cancer Lett. 2016;371:354-365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 53]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
110.  Yu S, Li Y, Liao Z, Wang Z, Qian L, Zhao J, Zong H, Kang B, Zou WB, Chen K, He X, Meng Z, Chen Z, Huang S, Wang P. Plasma extracellular vesicle long RNA profiling identifies a diagnostic signature for the detection of pancreatic ductal adenocarcinoma. Gut. 2020;69:540-550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 106]  [Article Influence: 26.5]  [Reference Citation Analysis (0)]
111.  Rong Z, Xu J, Shi S, Tan Z, Meng Q, Hua J, Liu J, Zhang B, Wang W, Yu X, Liang C. Circular RNA in pancreatic cancer: a novel avenue for the roles of diagnosis and treatment. Theranostics. 2021;11:2755-2769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
112.  Hogendorf P, Durczyński A, Skulimowski A, Kumor A, Poznańska G, Strzelczyk J. Neutrophil Gelatinase-Associated Lipocalin (NGAL) concentration in urine is superior to CA19-9 and Ca 125 in differentiation of pancreatic mass: Preliminary report. Cancer Biomark. 2016;16:537-543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
113.  Park WG, Li L, Appana S, Wei W, Stello K, Andersen DK, Hughes SJ, Whitcomb DC, Brand RE, Yadav D, Habtezion A; Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer. Unique circulating immune signatures for recurrent acute pancreatitis, chronic pancreatitis and pancreatic cancer: A pilot study of these conditions with and without diabetes. Pancreatology. 2020;20:51-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
114.  Lee B, Gibbs P. Inflammation, Biomarkers and Immuno-Oncology Pathways in Pancreatic Cancer. J Pers Med. 2019;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
115.  Brosens LA, Hackeng WM, Offerhaus GJ, Hruban RH, Wood LD. Pancreatic adenocarcinoma pathology: changing "landscape". J Gastrointest Oncol. 2015;6:358-374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 31]  [Reference Citation Analysis (0)]
116.  Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, Bignell GR, Bolli N, Borg A, Børresen-Dale AL, Boyault S, Burkhardt B, Butler AP, Caldas C, Davies HR, Desmedt C, Eils R, Eyfjörd JE, Foekens JA, Greaves M, Hosoda F, Hutter B, Ilicic T, Imbeaud S, Imielinski M, Jäger N, Jones DT, Jones D, Knappskog S, Kool M, Lakhani SR, López-Otín C, Martin S, Munshi NC, Nakamura H, Northcott PA, Pajic M, Papaemmanuil E, Paradiso A, Pearson JV, Puente XS, Raine K, Ramakrishna M, Richardson AL, Richter J, Rosenstiel P, Schlesner M, Schumacher TN, Span PN, Teague JW, Totoki Y, Tutt AN, Valdés-Mas R, van Buuren MM, van 't Veer L, Vincent-Salomon A, Waddell N, Yates LR; Australian Pancreatic Cancer Genome Initiative;  ICGC Breast Cancer Consortium;  ICGC MMML-Seq Consortium;  ICGC PedBrain, Zucman-Rossi J, Futreal PA, McDermott U, Lichter P, Meyerson M, Grimmond SM, Siebert R, Campo E, Shibata T, Pfister SM, Campbell PJ, Stratton MR. Signatures of mutational processes in human cancer. Nature. 2013;500:415-421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7533]  [Cited by in F6Publishing: 6777]  [Article Influence: 616.1]  [Reference Citation Analysis (1)]
117.  Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, Porta-Pardo E, Gao GF, Plaisier CL, Eddy JA, Ziv E, Culhane AC, Paull EO, Sivakumar IKA, Gentles AJ, Malhotra R, Farshidfar F, Colaprico A, Parker JS, Mose LE, Vo NS, Liu J, Liu Y, Rader J, Dhankani V, Reynolds SM, Bowlby R, Califano A, Cherniack AD, Anastassiou D, Bedognetti D, Mokrab Y, Newman AM, Rao A, Chen K, Krasnitz A, Hu H, Malta TM, Noushmehr H, Pedamallu CS, Bullman S, Ojesina AI, Lamb A, Zhou W, Shen H, Choueiri TK, Weinstein JN, Guinney J, Saltz J, Holt RA, Rabkin CS; Cancer Genome Atlas Research Network, Lazar AJ, Serody JS, Demicco EG, Disis ML, Vincent BG, Shmulevich I. The Immune Landscape of Cancer. Immunity. 2018;48:812-830.e14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3468]  [Cited by in F6Publishing: 3219]  [Article Influence: 536.5]  [Reference Citation Analysis (0)]
118.  Tewari N, Zaitoun AM, Arora A, Madhusudan S, Ilyas M, Lobo DN. The presence of tumour-associated lymphocytes confers a good prognosis in pancreatic ductal adenocarcinoma: an immunohistochemical study of tissue microarrays. BMC Cancer. 2013;13:436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 51]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
119.  Ryschich E, Nötzel T, Hinz U, Autschbach F, Ferguson J, Simon I, Weitz J, Fröhlich B, Klar E, Büchler MW, Schmidt J. Control of T-cell-mediated immune response by HLA class I in human pancreatic carcinoma. Clin Cancer Res. 2005;11:498-504.  [PubMed]  [DOI]  [Cited in This Article: ]
120.  Di Federico A, Mosca M, Pagani R, Carloni R, Frega G, De Giglio A, Rizzo A, Ricci D, Tavolari S, Di Marco M, Palloni A, Brandi G. Immunotherapy in Pancreatic Cancer: Why Do We Keep Failing? Cancers (Basel). 2022;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 22]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
121.  Karamitopoulou E, Andreou A, Pahud de Mortanges A, Tinguely M, Gloor B, Perren A. PD-1/PD-L1-Associated Immunoarchitectural Patterns Stratify Pancreatic Cancer Patients into Prognostic/Predictive Subgroups. Cancer Immunol Res. 2021;9:1439-1450.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
122.  Fernández-Carballo BL, Broger T, Wyss R, Banaei N, Denkinger CM. Toward the Development of a Circulating Free DNA-Based In Vitro Diagnostic Test for Infectious Diseases: a Review of Evidence for Tuberculosis. J Clin Microbiol. 2019;57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 53]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
123.  Sitek B, Sipos B, Alkatout I, Poschmann G, Stephan C, Schulenborg T, Marcus K, Lüttges J, Dittert DD, Baretton G, Schmiegel W, Hahn SA, Klöppel G, Meyer HE, Stühler K. Analysis of the pancreatic tumor progression by a quantitative proteomic approach and immunhistochemical validation. J Proteome Res. 2009;8:1647-1656.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 60]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]