Next Article in Journal
Stereotactic Body Radiotherapy for Renal Cell Carcinoma—A Review of Use in the Primary, Cytoreductive and Oligometastatic Settings
Previous Article in Journal
Demographic and Clinical Characteristics of Malignant Solitary Fibrous Tumors: A SEER Database Analysis
Previous Article in Special Issue
Circulating Multiple Myeloma Cells (CMMCs) as Prognostic and Predictive Markers in Multiple Myeloma and Smouldering MM Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Evolution of Liquid Biopsies for Detecting Pancreatic Cancer

1
Walter & Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
2
Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
3
Department of Medical Education, Melbourne Medical School, Parkville, VIC 3052, Australia
4
Western Health, Footscray, VIC 3011, Australia
5
Peter MacCallum Cancer Centre, Parkville, VIC 3052, Australia
6
Northern Health, Epping, VIC 3076, Australia
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(19), 3335; https://doi.org/10.3390/cancers16193335 (registering DOI)
Submission received: 6 September 2024 / Revised: 26 September 2024 / Accepted: 26 September 2024 / Published: 29 September 2024
(This article belongs to the Special Issue Liquid Biopsy in Cancer 2.0)

Abstract

:

Simple Summary

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy characterised by late diagnosis and poor prognosis, with current diagnostic and prognostic strategies proving insufficient. Liquid biopsy techniques, including circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), exosomes, and proteomics, present potential avenues for enhancing PDAC diagnosis, prognosis, and management. A review of literature from 2019 to 2024 identified 49 relevant studies focusing on these biomarkers. Investigations into ctDNA, particularly in detecting mutant KRAS, have shown some diagnostic and prognostic potential, though its efficacy in early-stage disease remains limited. CTCs, whilst highly specific, exhibit similarly restricted diagnostic utility in early disease, and studies have yielded inconsistent prognostic findings. Exosomal research identifies diverse biomarkers with promising diagnostic and prognostic value. Importantly, proteomics demonstrates superior accuracy in PDAC diagnosis, including early-stage detection, and significant prognostic capacity, especially when combined with CA19-9 analysis. This review highlights the significance of continued research into liquid biopsy techniques to improve PDAC management and outcomes.

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy characterised by late diagnosis and poor prognosis. Despite advancements, current diagnostic and prognostic strategies remain limited. Liquid biopsy techniques, including circulating tumour DNA (ctDNA), circulating tumour cells (CTCs), circulating tumour exosomes, and proteomics, offer potential solutions to improve PDAC diagnosis, prognostication, and management. A systematic search of Ovid MEDLINE identified studies published between 2019 and 2024, focusing on liquid biopsy biomarkers for PDAC. A total of 49 articles were included. ctDNA research shows some promise in diagnosing and prognosticating PDAC, especially through detecting mutant KRAS in minimal residual disease assays. CTC analyses had low sensitivity for early-stage PDAC and inconsistent prognostic results across subpopulations. Exosomal studies revealed diverse biomarkers with some diagnostic and prognostic potential. Proteomics, although relatively novel, has demonstrated superior accuracy in PDAC diagnosis, including early detection, and notable prognostic capacity. Proteomics combined with CA19-9 analysis has shown the most promising results to date. An update on multi-cancer early detection testing, given its significance for population screening, is also briefly discussed. Liquid biopsy techniques offer promising avenues for improving PDAC diagnosis, prognostication, and management. In particular, proteomics shows considerable potential, yet further research is needed to validate existing findings and comprehensively explore the proteome using an unbiased approach.

1. Introduction

Pancreatic cancer is a devastating malignancy of concern due to its late diagnosis and high mortality [1]. Pancreatic ductal adenocarcinoma (PDAC) accounts for over 85% of pancreatic malignancies [2] and has a terrible prognosis due to its aggressiveness and treatment-refractory nature [3]. This is reflected by its dismal overall 5-year survival rate of 12.5% in Australia [4], and a median survival rate of approximately 7 months after diagnosis [5]. Unlike most other malignancies, both the incidence and mortality rate of PDAC continue to rise, and it is expected to become the second leading cause of cancer-related death worldwide by 2030 [6]. Thus, significant improvements in patient diagnosis and management are urgently needed.
Diagnosis of PDAC typically occurs late in the disease course, with 80% of patients reporting unresectable, locally advanced, or metastatic disease at diagnosis [7,8]. Surgical excision remains the single most effective, and only potentially curative treatment option for PDAC. Patient prognosis is inversely correlated with tumour stage, with a 5-year survival rate as high as 80.4% for stage I disease [8]. However, most patients present with advanced disease and hence suffer a worse prognosis [9]. This highlights the need to improve early-stage PDAC diagnostics, ideally detecting disease through asymptomatic screening. However, due to the overall low incidence of PDAC, general population-based screening is currently not recommended [10]. Yet, there is a window of opportunity for earlier diagnosis in asymptomatic patients, particularly in the higher-risk groups such as those with hereditary pancreatitis, chronic pancreatitis, a strong family history of PDAC, and late new-onset diabetes [11]. Despite concerted efforts, there remains no validated diagnostic or screening test that is ready for large-scale early detection of PDAC.
In the current treatment paradigm for PDAC, chemotherapy regimen selection typically follows institutional evidence-based guidelines without incorporating the use of predictive or prognostic biomarkers to guide clinical decision-making [12]. Carbohydrate antigen 19-9 (CA19-9) is presently the only biomarker utilised for PDAC prognostication, but has notable limitations, failing to diagnose 35% of patients with resectable PDAC [13]. Its implementation as a tool for screening and diagnosis is impeded by low sensitivity of approximately 80%, with poorer performance for earlier stage disease [14], and limited specificity of approximately 82% due to elevation in inflammatory conditions, biliary pathologies, and non-pancreatic malignancies [15]. With regards to PDAC prognostication, several surgical classification guidelines have recommended incorporating CA19-9 into the selection paradigm for borderline resectable pancreatic cancers (BRPC). Such guidelines utilise CA19-9 cut-offs ranging from 150 to 500 U/mL to select candidates with ‘high-risk’ characteristics of metastatic disease for pre-operative neoadjuvant chemotherapy approach prior to resection [16,17,18]. However, CA19-9 findings are generally only interpreted in combination with other investigations, such as imaging, and hence there is ongoing need for more reliable biomarkers for PDAC screening, diagnosis, and prognostication [17,19,20].
There is now growing interest in liquid biopsies as a means to survey tumour-derived biomarkers in peripheral blood, urine, and other biological fluids, such as peritoneal washings, in order to remotely assess disease burden. These methods boast the benefits of being non-invasive, relatively simple, and can safely be repeated multiple times to obtain longitudinal data [21]. This approach addresses the limitations of detecting early-stage PDAC, including diagnosing small tumours that are undetectable by radiological imaging and that are too small to biopsy by fine-needle aspiration (FNA). Liquid biopsies also overcome limitations of restricted tumour tissue availability and hurdles in obtaining specimens suitable for molecular analysis. Due to the hypocellular and stromal-rich nature of PDAC tumours [22], technical issues of insufficient tumour sampling or limited tumour cellularity are frequently encountered when attempting to perform molecular profiling from endoscopically obtained FNA cell block specimens [23].
Identification of novel biomarkers for PDAC holds immense potential to not only improve early detection and diagnosis, but also to support prognostication and guide patient management [24]. The applications of such techniques have been demonstrated for other cancer types, such as lung and breast cancer [25,26], but clinical applications in PDAC have only recently emerged. Data now demonstrate the feasibility of liquid biopsy techniques to guide adjuvant chemotherapy selection for early-stage PDAC, [27] as well as serial biopsy monitoring for predicting treatment response and prognosis [28].
Biomarker technologies investigated for the purpose of liquid biopsy of PDAC include circulating tumour DNA (ctDNA), circulating tumour cells (CTCs), and circulating tumour exosomes [29]. More recently, proteomics has emerged as a promising alternative liquid biopsy option, but remains poorly characterised in pancreatic cancer. This review will examine each of these four technologies in turn, as shown in Figure 1, scrutinising recent developments and comparing their clinical utility in PDAC.

2. Methods

A systematic search of online database Ovid MEDLINE was performed in February 2024. Search keywords and Medical Subject Headings were related to pancreatic cancer and liquid biopsy techniques of interest to this study. The Boolean ‘OR’ operator was used to group related terms, and these groups were then combined using the Boolean ‘AND’ operator. This search returned 3147 articles.
Limits were applied to the initial search strategy to meet the following inclusion criteria: (1) investigated adults only (defined as age ≥ 19 years on Ovid); (2) article available in English; and (3) article published 2019–present, given a major literature review of the topic was published in 2019 [29]. This limited the search results to 156 papers, which were then individually appraised for relevance. Papers were excluded based on the following exclusion criteria: (1) examination of non-PDAC pancreatic cancer; (2) examination of tissue-sampled biomarkers instead of peripheral fluids; (3) failure to investigate screening, diagnostic, prognostication, or therapeutic monitoring applications of the biomarker; and (4) case reports, letters, editorials, or reviews. The final number of accepted articles was 49. The PRISMA flow diagram outlining the literature search is depicted in Figure 2. The literature search strategy (prior to manual application of exclusion criteria) is described in Figure A1.

3. Discussion

3.1. Circulating Tumour DNA

Circulating tumour DNA (ctDNA) is a fraction of cell-free DNA (cfDNA) consisting of fragmented tumour-derived DNA shed from primary tumours or metastases and found in plasma and urine [30]. Methods of ctDNA analysis generally involve either sequencing of tumour-specific mutations or detecting aberrant DNA methylation patterns [31]. Additionally, there are two broad approaches for investigating ctDNA depending on whether the genetic aberrations present in the primary tumour are known or not, termed tumour-informed and tumour-agnostic techniques, respectively. In tumour-agnostic studies, ctDNA samples are analysed without prior examination of tumour tissue, and therefore target frequently mutated genes such as KRAS, or epigenetic changes such as hypermethylation in the promotor regions of tumour suppressor genes [32]. This approach is generally used in the context of cancer screening, and molecular diagnostic testing of patients with unresectable solid tumours, with next generation sequencing (NGS)-based assays commonly employed in such studies [33]. Conversely, tumour-informed approaches utilise sequencing data obtained from patient tumour tissue to create personalised gene panels that can be applied to ctDNA analysis [34]. Applications include detection of minimal residual disease or recurrence following curative-intent surgery, and monitoring of patient-specific therapeutic responses. Methods that adopt this tumour-informed approach include targeted sequencing platforms modified with molecular barcoding methods and error-suppression algorithms, such as Safe-SeqS, as well as polymerase chain reaction (PCR)-based platforms, such as droplet digital PCR (ddPCR) [35]. Tumour-informed studies have intrinsically higher sensitivity in detecting disease due to their application in only patients with previously known tumours [34]. Overall, there is much ongoing interest exploring the value of ctDNA analysis in guiding PDAC diagnosis, prognostication, and management. Details of the 22 identified studies pertaining to ctDNA analysis are outlined in Table 1.
The diagnostic utility of KRAS-mutated ctDNA in PDAC patient plasma has recently been explored by several groups, reporting varying sensitivities based on different assays. Among newly diagnosed patients, published sensitivities include 37% [36], 48% [37,38], 49% [39], and 62% [40]. Others have trialled panels of cancer-related genes and have described sensitivities of 38% [41], 44% [42], 45% [43], 50% [44], 66% [45], 68% [46], and 75% [47]. Additionally, Terasawa et al. examined ctDNA in PDAC patient urine and reported a sensitivity of 48% whilst also identifying that renal function may act as a potential confounder [38]. These studies frequently identified mutated KRAS and TP53 as the most common ctDNA fragments in PDAC patient plasma, consistent with their role in PDAC oncogenesis [48], yet these low reported sensitivities somewhat undermine the feasibility of ctDNA-based diagnostic testing. Considering MacGregor-Das et al. identified KRAS ctDNA-positivity in just 31% of patients with stage I/II disease [36], whether ctDNA alone will contribute a major role to asymptomatic PDAC screening in the future is also yet to be determined. Furthermore, the positive predictive value of mutational ctDNA analysis is hampered by false positive results arising from multiple sources. KRAS-mutated ctDNA is carried by some healthy individuals, as noted in the preceding study [36], which is consistent with prior understanding that non-cancerous circulating lymphoid and myeloid cells can accumulate confounding somatic mutations [49]. False positive results may also arise ex vivo due to sequencing read errors [50] and inadvertent chemical modification of ctDNA, such as by cytosine deamination [36].
As an alternative means for diagnostic testing, ctDNA hypermethylation patterns have been described for several genes. Pietrasz et al. identified HOXD8 and POU4F1 hypermethylation had a sensitivity of 56.8% in a large cohort of 372 patients with metastatic PDAC [51]. Another group found SPARC and NPTX2 hypermethylation can distinguish PDAC from healthy controls and patients with chronic pancreatitis [52]. Alternatively, ZNF154 has been reported to discern cancer patients, including those with pancreatic, colorectal, and liver malignancies, from healthy controls [53]; however, such testing inherits intrinsic limitations to specificity. Other studies have combined methylation analysis with other metrics to improve test performance. For example, Shinjo et al. found the sensitivity of a five gene panel containing ADAMTS2, HOXA1, PCDH10, SEMA5A, and SPSB4 improved from 49% to 68% when combined with testing for KRAS-mutated ctDNA [54]. Similarly, Fujimoto et al. reported an increase in sensitivity from 50.9% to 85.5% for a RUNX3-based assay, once combined with CA19-9 measurement [55]. However, despite these increases, the sensitivities of such tests remain generally low and thus have limited clinical utility.
Perhaps more promising is the prognostic potential of ctDNA detection. Several groups found significant associations between mutant KRAS–ctDNA positivity and inferior prognosis across the spectrum of disease, including pre-operative patients, those with residual disease post-resection, and those with metastatic disease. Specifically, positive mutant KRAS–ctDNA status has been associated with earlier tumour recurrence, greater metastatic burden, and shorter median progression-free survival (mPFS) and median overall survival (mOS) [37,39,40,41,56,57,58]. These findings were also mirrored in gene panel studies [43,44,45,46,47]. Only one group reported no association between pre-operative KRAS–ctDNA status and overall prognosis [59]. There are considerably fewer publications exploring methylation-based prognostication, yet they largely support these mutational studies. Singh et al. found SPARC and NPTX2 hypermethylation correlate with disease stage and poorer survival [52], whilst Pietrasz et al. identified HOXD8 and POU4F1 as independent prognostic markers for mPFS and mOS [51], and Wang et al. found that total unfractionated cfDNA also correlates with poorer patient prognosis [60]. Overall, these studies support a prognostic role for ctDNA analysis in PDAC patients.
Furthermore, some research now links ctDNA testing to predictive therapeutic utility. Decreases in mutant KRAS–ctDNA titres within six months of commencing first-line palliative chemotherapeutics has been correlated with better treatment response [59]. Similarly, Bachet et al. report decreases in ctDNA among patients receiving eryaspase therapy correlates with better drug efficacy [46]. Wei et al. also report that increases and decreases in ctDNA methylation for a variety of genes correlate with chemotherapy resistance and responsiveness, respectively [45]. These early studies show the potential for ctDNA analysis to guide patient management via selection of effective therapeutics; however, further studies in larger cohorts will be required for clinical validation. Furthermore, relatively few studies utilising tumour-informed ctDNA analysis have been published, and such studies would further support these findings.
Table 1. Characteristics of ctDNA studies identified for analysis.
Table 1. Characteristics of ctDNA studies identified for analysis.
CitationPatientsApplications ExploredMethodology
NDisease DescriptorStudy AimDiagnosis (Sensitivity, Specificity)PrognosticPredictiveAgnostic/
Informed
Broad MethodDetection TechnologyGene(s) Examined
Bachet et al., 2020 [46]113Stage IVTo evaluate the prognostic and predictive value of ctDNA from plasma samples during a randomised phase II trial which has assessed eryaspase efficacy in patients with advanced PDAC.Yes
(68.1, NR)
YesYesAgnosticMutationAmpliSeq colon and lung cancer panel V2 (NGS)22
Cheng et al., 2020 [58]210Stage III–IVTo evaluate the KRAS mutation status in ctDNA and circulating T cell subsets in a cohort of advanced pancreatic cancer patients.NoYesNoAgnosticMutationddPCRKRAS
Chung et al., 2021 [42]77Stage IVTo characterise the mutational landscape of patients with metastatic PDAC who received blood-based molecular profiling.Yes
(44.1, NR)
NoNoAgnosticMutationGuardant-360 (NGS)83
Fujimoto et al., 2021 [55]55Stage I–IVTo evaluate the sensitivity and specificity of serum DNA testing of methylated RUNX3 by the CORD assay for the detection of PDAC.Yes
(50.9, 93.5)
NoNoAgnosticHMCORD assayRUNX3
Groot et al., 2019 [39]59Pre- and post-operativeTo perform analytical and clinical validation of a KRAS ctDNA assay in a certified clinical laboratory.Yes
(49.2, NR)
YesNoAgnosticMutationddPCRKRAS
Guo et al., 2020 [41]113Pre-operativeTo explore the clinical value of hotspot mutations in resectable PDAC patients.Yes
(38.1, NR)
YesNoAgnosticMutationNGS50
Hussung et al., 2021 [37]25Post-operativeTo identify associations between mutant cfKRAS and CA19-9 dynamics and clinical outcome post resection.Yes
(48.0, NR)
YesNoAgnosticMutationddPCRKRAS
Lee et al., 2019 [40]42Pre-operativeTo evaluate the feasibility and clinical utility of ctDNA analysis to inform adjuvant therapy decision making.Yes
(62.2, NR)
YesNoInformedMutationSafe-SeqS assay (PCR)KRAS
Li et al., 2021 [56]105Pre-operativeTo establish a scoring system for preoperative screening of resectable PDAC patients.NoYesNoAgnosticMutationqPCRKRAS
Macgregor-Das et al., 2020 [36]67Screening and pre-operativeTo utilise enzymatic pretreatment of plasma DNA followed by digital NGS to detect hotspot mutations in KRAS and GNAS in patients with pancreatic cancer.Yes
(36.5, 92.0)
NoNoAgnosticMutationDigital NGSKRAS
GNAS
Miller et al., 2021 [53]17Stage I–IVTo assess the effectiveness of hypermethylation at the CpG island of ZNF154 for use in a blood-based cancer detection assay.Yes
(NR, NR)
NoNoAgnosticHM ddPCRZNF154
Patel et al., 2019 [47]112Stage I–IVTo assess the genomic landscape of ctDNA in patients with PDAC, using clinical-grade NGS to investigate the clinical implications.Yes
(69.6, NR)
YesNoAgnosticMutationNGS54–73
Pietrasz et al., 2022 [51]372Stage IV; CTx-naïveTo determine whether ctDNA is an independent factor for the prognostication of metastatic PDAC.Yes
(56.8, NR)
YesNoAgnosticHMddPCRHOXD8
POU4F1
Shinjo et al., 2020 [54]47Stage I–IVTo identify effective DNA methylation markers for the diagnosis of PDAC.Yes
(49.0, 86.0)
NoNoInformedHMMBD-ddPCRHOXA1
PCDH10
ADAMTS2
SEMA5A
SPSB4
Singh et al., 2020 [52]65Stage I–IVTo conduct absolute quantification of methylation in SPARC, UCHL1, PENK, and NPTX2 genes and assess the respective methylation load for their ability to perform as non-invasive differentiating and prognostic marker(s) for PDAC.Yes
(NR, NR)
YesNoAgnosticHMqPCRSPARC
UCHL1
PENK
NPTX2
Strijker et al., 2020 [43]58Stage IV; CTx-naïveTo evaluate the capability of targeted sequencing using a custom pancreatobiliary NGS panel and of ddPCR to detect ctDNA in metastatic PDAC.Yes
(44.8, NR)
YesNoAgnosticMutationNGS8
Sugimori et al., 2020 [57]21Stage IVTo assess the dynamics of ctDNA in patients with advanced PDAC undergoing chemotherapy using dPCR.NoYesNoAgnosticMutationdPCRKRAS
Terasawa et al., 2019 [38]56Stage II–IVTo analyse KRAS mutations in urine to investigate the potential of urine liquid biopsy in PDAC.Yes
(48.2, NR)
NoNoAgnosticMutationddPCRKRAS
Uesato et al., 2020 [44]104Stage IVTo evaluate circulating tumor DNA as a tumor biomarker to prognosticate pancreatic cancer.Yes
(50.0, NR)
YesNoAgnosticMutationOncomine colon cfDNA assay (NGS)14
Wang et al., 2021 [60]97Pre-operativeTo evaluate the role of cfDNA in PDAC of the pancreatic head. NoYesNo-Total cfDNAqPCR-
Watanabe et al., 2019 [59]78Pre- and post- CTx and/or surgeryTo evaluate the significance of sequentially assessing KRAS ctDNA levels through longitudinal monitoring.NoYesYesInformedMutationddPCRKRAS
Wei et al., 2019 [45]38Receiving palliative CTxTo explore the application of cfDNA profiling in monitoring tumor burden in patients with PDAC.Yes
(65.8, 100)
YesYesAgnosticMutationNGS560
Notes: N denotes number of PDAC patients included in study only. Most appropriate descriptor of disease has been applied, including AJCC (8th) disease stage or resection state. Diagnostic sensitivity is reported relative to healthy controls, without stratification by disease stage, and without combination with other metrics (e.g., CA19-9). Genes examined presented as names if ≤5 reported, or number of genes in panel if >5 reported. All numbers reported to three significant figures, wherever possible. Abbreviations: ctDNA, circulating tumour DNA; cfDNA, cell-free DNA; NR, not reported (or unable to be deconvoluted from reported data); HM, hyper-methylation; NGS, next-generation sequencing; PCR, polymerase chain reaction; dPCR, digital PCR; ddPCR, droplet digital PCR; CORD, combined restriction digital PCR; qPCR, quantitative PCR; MBD-ddPCR, methyl-CpG binding ddPCR; CTx, chemotherapy.

3.2. Circulating Tumour Cells

Circulating tumour cells (CTCs) are malignant cells that can be isolated from the peripheral blood of cancer patients, providing some measure of tumour burden [61]. Several recent studies have correlated the presence and/or quantity of CTCs with clinical data in PDAC. These studies have historically analysed the total population of a patient’s CTCs, but, more recently, research is emerging exploring CTC subpopulations. Details of the eight identified studies pertaining to CTC analysis are delineated in Table 2.
Studies investigating the diagnostic utility of assessing total CTCs have been somewhat discouraging. A large study of 106 PDAC patients reported a trend of increasing sensitivity with disease stage, with CTC-positivity rates ranging from just 37.5% for stage II to 100% for stage IV patients [62]. Discounting disease stage, several groups have published findings of overall CTC positivity in 75% [63], 76% [64], 77% [65], and 79% [66] of PDAC patients, whilst Park et al. have reported 33% positivity among patients post-resection [67]. Each of these studies also explored the prognostication capacity of total CTC analysis, with variable findings. Many groups correlated CTC-positivity with poorer prognosis, including increased recurrence risk and shorter mPFS [62,63,64,66,67], whilst Semaan et al. found no correlation between total CTC count and any clinicopathological variables in their study of 74 PDAC patients [65].
More recently, these groups and others have identified subpopulations of CTCs related to epithelial–mesenchymal transition (EMT) phenotypes. Three subpopulations are commonly identified based on expression of epithelial markers (generally cytokeratin and EpCAM), mesenchymal markers (generally vimentin and Twist), or both (termed ‘mixed’ CTCs). With respect to diagnostic testing, detection of mesenchymal CTCs in conjunction with CA19-9 testing was found to hold some diagnostic potential, with an area under the receiver operator curve (AUROC) of 0.968 [64]. Similarly, isolation of an unrelated CTC subpopulation based on folate receptor expression among patients with pancreatic malignancy achieved an AUROC of 0.944 when combined with CA19-9 [68]. Despite these improvements in diagnostic potential, explorations of subpopulation prognostic capacity have yielded variable results. Some groups have found that a higher CTC proportion exhibiting a mixed phenotype is correlated with advanced disease stage [66], earlier recurrence, and worse mPFS and mOS [65]. Alternatively, Xing et al. reported that both mixed and mesenchymal CTC-positivity were associated with shorter mPFS [62], whilst Zhu et al. identified that only mesenchymal CTCs were linked to a worse prognosis [63]. These data broadly align with prior research that has established CTCs with a mesenchymal or mixed phenotype show greater invasiveness [69], and thus may be linked to a poorer prognosis. However, a small study of 21 patients by Gasparini et al. reported no significant relationship between mesenchymal CTCs and mPFS or mOS [70].
Considered together, these data reveal persistent limitations in the utility of CTC analysis for PDAC diagnosis and prognostication. In particular, low sensitivity in detecting earlier stage disease [62] renders prospects of developing CTC-based screening impractical at this time. Moreover, conclusions reached by these studies in identifying clinicopathological correlates for prognostication exhibit stark inconsistencies. This lack of consensus hinders the development of reliable CTC-based testing due to lack of established thresholds or endpoints.
Variability in reported findings may be, in part, attributable to differences in study methodology. Briefly, there are two fundamental techniques for CTC isolation based on either biomolecule expression or physical properties, which are known to somewhat underestimate and overestimate results, respectively [29]. As such, some variability in conclusions drawn by the reviewed articles may be explained by differences in CTC detection techniques, given some studies utilised expression-based assays (such as immunomagnetic separation), whilst others used morphology-based assays (such as dielectrophoretic-field flow fractionation). By extension, the intrinsic inaccuracies of the testing modalities, coupled with the rarity and low yield of isolating CTCs from patient blood [71], render CTC-based test development difficult.
Table 2. Characteristics of CTC studies identified for analysis.
Table 2. Characteristics of CTC studies identified for analysis.
CitationPatientsApplications ExploredMethodology
NDisease DescriptorStudy AimDiagnosis
[AUROC] or (Sensitivity, Specificity)
PrognosticTotal/
Subpops
Biomolecule/PhysicalIsolation Technology
Cheng et al., 2020 [68]45 aStage I–IVTo investigate the diagnostic value of folate receptor-positive CTCs in distinguishing pancreatic cancer from benign pancreatic disease.Yes
[0.837]
NoFolate-receptor positiveBiomoleculeLT-PCR
Gasparini-Junior et al., 2019 [70]21Stage II–IVTo correlate the number of CTCs in the peripheral blood of patients with locally advanced or metastatic pancreatic tumors, and the protein expression involved in EMT in CTCs with clinical characteristics, mPFS, and mOS.NoYesTotal and EMT subpopsPhysicalISET
Park et al., 2021 [67]36Post-operativeTo determine whether the preoperative presence of CTCs is associated with the overall survival and recurrence-free survival in patients with PDAC.Yes
(33.3, NR)
YesTotal onlyPhysicalCD-PRIME
Semaan et al., 2021 [65]74Stage I–IVTo perform comprehensive phenotypic characterisation of CTCs and their clinical significance in a longitudinal cohort of PDAC patients.Yes
(76.8, NR)
YesTotal and EMT subpopsPhysicalDEP-FFF
Wei at al., 2019 [64]100Stage I–IVTo determine whether cell-surface vimentin could be a biomarker to isolate CTCs in PDAC.Yes
(76.0, 97.4)
YesTotal and EMT subpopsBiomoleculeCytoQuest CR
Xing et al., 2022 [62]106Stage II–IVTo explore the relationship between CTCs or T lymphocyte subsets and prognosis in patients with pancreatic cancer.Yes
(75.5, NR)
YesTotal and EMT subpopsPhysicalCanPatrol CTC
Zhao et al., 2019 [66]107Stage I–IVTo evaluate the clinical properties of three CTC subpopulations undergoing EMT in PDAC patients.Yes
(78.5, NR)
YesTotal and EMT subpopsPhysicalCanPatrol CTC
Zhu et al., 2021 [63]40Pre-operativeTo determine the prognostic significance of CTCs expressing Krüppel-like factor 8 and vimentin in pancreatic cancer.Yes
(75.0, NR)
YesTotal and EMT subpopsBiomoleculeImmuno-magnetic separation
Notes: N denotes number of PDAC patients included in study only. Most appropriate descriptor of disease has been applied, including AJCC (8th) disease stage or resection state. Diagnostic performance is reported as AUROC where possible, or sensitivity and specificity; reported relative to healthy controls, without stratification by disease stage, and without combination with other metrics (e.g., CA19-9). All numbers reported to three significant figures, wherever possible. Abbreviations: CTC, circulating tumour cell; AUROC, area under receiver operator curve; subpops, subpopulations; EMT, epithelial–mesenchymal transition; NR, not reported (or unable to be deconvoluted from reported data); PCR, polymerase chain reaction; LT-PCR, ligand-targeted PCR; ISET, isolation by size of tumour cells; DEP-FFF, dielectrophoretic field-flow fractionation. a Study population included 4% patients with intraductal papillary mucinous neoplasms, and data could not be deconvoluted to examine PDAC only.

3.3. Circulating Tumour Exosomes

Circulating tumour exosomes are extracellular vesicles released by cells, including cancer cells, into peripheral blood [72]. The content of an exosome reflects that of its parent cell, and typically includes many bioactive molecules such as nucleic acids, proteins, and lipids [29]. Consequently, recent studies have sought to explore the diagnostic and prognostic potential of exosome analysis in PDAC. Details of the nine identified studies pertaining to exosome analysis are outlined in Table 3.
Recent developments have examined several types of nucleic acids in circulating tumour exosomes. With respect to PDAC diagnostics, expression of long non-coding RNA HULC has been identified as being significantly elevated in patient plasma compared to healthy controls and intraductal papillary mucinous neoplasm (IPMN) patients [73]. Similarly, Kitagawa et al. reported messenger RNAs WASF2 and ARF6, and small nucleolar RNAs SNORA74A and SNORA25, can each distinguish pancreatic cancer patients from healthy controls with an AUROC of >0.9, including for patients with stage I/II disease [74]. Furthermore, a suite of microRNAs capable of distinguishing PDAC from related pathologies has been published by Vicentini et al., with nine, twenty, and twenty-two miRNAs found to differ significantly in PDAC, compared to chronic pancreatitis, IPMN, and ampulla of Vater carcinoma, respectively. Moreover, regarding prognostication, this group also identified a signature of 11 miRNAs able to differentiate localised PDAC from metastatic disease [75]. Thus, nucleic acid analysis holds potential for PDAC diagnosis and prognostics; however, these studies cumulatively examined just 102 patients, and thus validation in larger cohorts is warranted. In like manner, one study investigating exosomal lipids has proved similar merit, identifying approximately 270 lipids that are significantly dysregulated in pancreatic cancer patients, and that specifically lysophosphatidylcholine (22:0), phosphatidylcholine (P-14:0/22:2), and phosphatidylethanolamine (16:0/18:1) are significantly correlated with tumour stage and mOS [76]. However, this study suffers the similar limitation of examining just 22 patients.
Explorations of exosomal protein expression have yielded some promising results, and in larger cohorts. Diagnostic studies by Wei et al. identified exosomal Eph receptor A2 (EphA2) levels were significantly elevated among 244 pancreatic cancer patients. Indeed, AUROC values for serum exo-EphA2 diagnostic testing were 0.94 and 0.92 against healthy controls and benign pancreatic disease, respectively. These values decreased slightly when examining early PDAC (stages I/II) to 0.92 and 0.90, respectively, yet combining exo-EphA2 with CA19-9 measurement then raised AUROC to 0.96 against healthy controls [77,78]. These authors also found serum exo-EphA2 levels were significantly higher among patients with advanced disease (stages III/IV) compared to those with earlier stage disease, providing some indication of prognostic value. Whilst encouraging, these data would benefit from replication by an independent research group. Xiao et al. achieved similar diagnostic efficacy using a panel of serum exosomal glypican-1, exosomal CD82, and CA19-9, publishing an AUROC of 0.942 [79]. Conversely, another group examining exosomal c-Met expression reported significantly higher expression levels among PDAC patients compared to benign disease, but poor diagnostic capacity with a sensitivity of 70% and specificity of 85% [80]. Regarding prognostication, these authors did, however, note exosomal c-Met-positivity and PD-L1-positivity correlated with shorter mOS after surgery. Interestingly, Giampieri et al. identified that a higher quantity of exosomal EpCAM was associated with poorer response to palliative chemotherapy and shorter mOS [81]. This is seemingly at odds with recent research into CTC-based PDAC prognostics, as discussed previously, whereby several groups identified CTCs adopting a mesenchymal EpCAM-negative phenotype were associated with poorer prognosis, consistent with the invasive characteristics associated with mesenchymal tumour cells [69]. However, this group also identified increases in EpCAM expression during chemotherapy were instead associated with longer mPFS [81]. Such inconsistencies may be, in part, explained by the small sample size of 19 for this study. Overall, much like nucleic acid- and lipid-based research, exosomal proteins analysis offers generally good test characteristics for the diagnosis and prognostication of PDAC. However, validation of the discussed findings must be conducted in larger cohorts, with a goal of corroboration between multiple research groups for the studied disease markers.
Table 3. Characteristics of circulating tumour exosome studies identified for analysis.
Table 3. Characteristics of circulating tumour exosome studies identified for analysis.
CitationPatientsApplications ExploredMethodology
NDisease DescriptorStudy AimDiagnosis
[AUROC] or (Sensitivity, Specificity)
PrognosticBiomolecule ClassAnalysis TechniqueMolecule(s) Examined
Giampieri et al., 2019 [81]19Receiving palliative CTxTo analyse the contents of circulating exosomes in patients with pancreatic cancer who received palliative CTx.NoYesProteinELISA10
Kitagawa et al., 2019 [74]27Stage I–IIITo assess the utility of several serum mRNAs and snoRNAs as diagnostic markers for differentiating PDAC patients from control patients without pancreatic disease.Yes
[0.883] a
YesmRNA
snoRNA
RT-qPCR4 (mRNA)
5 (snoRNA)
Lux et al., 2019 [80]55Stage I–IVTo assess whether pancreatic carcinomas release exosomes which express c-Met and PD-L1, and whether the detection of such expression in serum has diagnostic or prognostic meaning for the affected patients.Yes
(70, 85)
YesProteinFlow cytometryc-Met
PD-L1
Takahashi et al., 2020 [73]20Stage II–IVTo identify lncRNAs involved in the EMT pathway and investigate their functional roles during PDAC cell invasion and migration.Yes
[0.92] b
YeslncRNAdPCRHULC
Tao et al., 2019 [76]22Post-operativeTo identify the possible prognostic or diagnostic metabolite biomarkers in the serum exosome of PDAC patients.Yes
(NR, NR)
YesLipidLC-DDA-MS270
Vicentini et al., 2020 [75]55Stage I–IVTo identify circulating miRNAs able to discriminate different histotypes of pancreatobiliary neoplasms.Yes
(NR, NR)
YesmiRNANanoString22
Wei et al., 2020 [77]40Stage I–IVTo understand the mechanism underlying pancreatic cancer metastasis to identify novel biomarkers.Yes
(NR, NR)
YesProteinELISAEphA2
Wei et al., 2020 [78]204Stage I–IVTo evaluate serum Exo-EphA2 as a potential diagnostic biomarker in pancreatic cancer.Yes
[0.94]
YesProteinELISAEphA2
Xiao et al., 2020 [79]27NRTo establish a simple and efficient standard method for the detection and analysis of exosomal GPC1 protein to explore screening value in Chinese patients with pancreatic cancer.Yes
[0.869] c
NoProteinFlow cytometryGPC1
CD82
Notes: N denotes number of PDAC patients included in study only. Most appropriate descriptor of disease has been applied, including AJCC (8th) disease stage or resection state. Diagnostic performance is reported as AUROC where possible, or sensitivity and specificity; reported relative to healthy controls, without stratification by disease stage, and without combination with other metrics (e.g., CA19-9). Molecule(s) examined presented as names if ≤5 reported, or number of molecules if >5 reported. All numbers reported to three significant figures, wherever possible. Abbreviations: AUROC, area under receiver operator curve; NR, not reported (or unable to be deconvoluted from reported data); RNA, ribonucleic acid; mRNA, messenger RNA; snoRNA, small nucleolar RNA; lncRNA, long non-coding RNA; miRNA, micro-RNA; ELISA, enzyme-linked immunosorbent assay; PCR, polymerase chain reaction; RT-qPCR, reverse transcription quantitative PCR, dPCR, digital PCR; LC-DDA-MS, liquid chromatography data-dependent acquisition mass spectrometry; CTx, chemotherapy. a AUROC reported as median of published values for each of nine independent markers. b AUROC reported as relative to both healthy controls and IPMN patients combined. c AUROC reported as mean of published values for two independent markers.

3.4. Proteomics

The application of free protein-based testing for PDAC is now an emerging field for liquid biopsies that boasts promising early results. Proteomic studies to date have mostly analysed the proteome of patient plasma and sera, enabling an in-depth dynamic assessment of how genes are expressed, and protein–protein interactions. Compared to targeted genomic analysis, which is a static assessment of mutational tumour DNA, the systemic large-scale analysis of proteins offers increased technical capability to detect rarer alterations, functional changes, as well as a better understanding of the nature of protein-drug interactions. Details of the 10 identified studies pertaining to analysis of circulating proteins are described in Table 4.
One large study of 401 PDAC patients by Kim et al. investigating the diagnostic capabilities of a 14-marker proteomics panel achieved AUROC values of 0.977, 0.953, and 0.928, for one exploratory and two independent validation cohorts, respectively [82]. This study involved many control populations, including healthy volunteers, patients with benign pancreatic lesions, and patients with other types of cancer. The diagnostic performance of this panel significantly outperformed CA19-9 alone, yet together these metrics were able to reach an impressive AUROC of 0.989. Similarly, Wu et al. identified PROZ and TNFRSF6B serum levels could significantly distinguish PDAC patients from healthy controls and benign pancreatic pathologies, achieving an AUROC of 0.971 overall and 0.913 for stage I disease, when combined with CA19-9 [83]. A PDAC risk prediction model, developed by Kartsonaki et al., based on known risk factors such as age, smoking, and alcohol consumption, significantly improved after factoring in serum concentrations of several proteins, including angiopoietin-2, monocyte chemotactic protein-3, interleukin-6, and interleukin-18, with a significant increase in weighted C-statistic from 0.85 to 0.99 [84]. Smaller studies have achieved varying diagnostic success. For example, one study of twenty-two patients identified a panel of seven peptides able to distinguish PDAC from healthy controls with 93% sensitivity [85], whilst another group identified a panel of eight proteins able to discriminate early PDAC from healthy individuals with an AUROC of 0.81–0.85 [86]. Comparably, Deutsch et al. examined the salivary proteome of fifteen male PDAC patients and found a panel of five proteins (cytokeratin-14, cytokeratin-16, cytokeratin-17, lactoperoxidase, and peptidyl-prolyl cis–trans isomerase B) that could discern patients from healthy controls with an AUROC of 0.91 [87].
With respect to proteomics based PDAC prognostication, few studies have yet been published, and these generally examine relatively small cohorts. Sahni et al. identified lower plasma levels of protein tyrosine phosphatases PTPRB and PTPRM were associated with poorer prognosis in a study of 12 patients [88], whilst Holm et al. identified a panel of 31 proteins, including several enzymes and complement proteins, which significantly differed between 21 PDAC patients with long or short mOS [89]. Similarly, elevated concentration of plasma fucosylated SERPINA1 has been significantly correlated with disease stage and shorter mOS [90]. Furthermore, Rittman et al. reported that serum concentration of 23 proteins, including MAEA, could effectively distinguish PDAC patients with early versus late recurrence. This group further identified MAEA expression is significantly elevated by several PDAC organoid lines, and that organoid-derived MAEA levels could similarly discriminate early and late recurrence [91]. Therefore, whilst the development of proteomics-based PDAC testing is relatively novel, early studies have showed incredible potential for diagnostic utility, particularly in conjunction with CA19-9 testing, having reported the highest AUROC values of all related publications in the past five years. However, in a similar vein to circulating tumour exosomes, validation and corroboration of research findings between groups remains to be seen and will be required moving forwards. Furthermore, several of these studies analysed only pre-determined candidate proteins, and comprehensive explorations of the proteome among large and diverse PDAC cohorts remain limited. Regarding prognostication, relatively little evidence yet exists, and, similarly, larger exploratory and validation studies are needed. The relative limitations and strengths of the four PDAC biomarker modalities discussed herein are summarised in Table 5.
Table 4. Characteristics of proteomics studies identified for analysis.
Table 4. Characteristics of proteomics studies identified for analysis.
CitationPatientsApplications ExploredMethodology
NDisease DescriptorStudy AimDiagnosis
[AUROC] or (Sensitivity, Specificity)
PrognosticCandidate OnlyAnalysis TechniqueProtein(s) Examined
Deutsch et al., 2020 [87]15Stage III–IVTo identify and develop an early detection assay for pancreatic cancer based on candidate biomarkers in oral fluids.Yes
[0.91]
NoNo—comp
(oral fluids)
DM-LCT-MSCytokeratin-14
Cytokeratin-16
Cytokeratin-17
Lacto-peroxidase
Peptidyl-prolyl cis–trans isomerase B
Duan et al., 2019 [85]22Stage I–IIITo identify differentially expressed peptides involved in pancreatic cancer as potential biomarkers.Yes
(93.0, 94.6)
NoNo—compLC-ESI-MS7
Holm et al., 2020 [89]21Stage I, II, IVTo analyse the serum proteome of a small cohort of PDAC patients and compare the protein expression between patients with short- and long-term survival, in order to discover proteins that could be of value as new candidates for prognostic biomarkers. NoYesNo—compUPLC-UDMS31
Kartsonaki et al., 2022 [84]610NRTo examine the prospective associations of >90 protein biomarkers with development of pancreatic cancer and to assess the extent to which they could help predict risk of a future diagnosis.Yes
(C-statistic) a
NoYesOlink—immuno-oncology panel92
Kim et al., 2021 [82]401Stage I–IVTo develop and validate a protein-based, multi-marker panel that provides superior PDAC detection abilities with sufficient diagnostic performance.Yes
[0.977]
NoYesMRM-MS14
Rittmann et al., 2021 [91]14Pre-operativeTo identify preoperative plasma protein biomarkers with the potential to predict early recurrence after resection of PDAC.NoYesYesOlink—several panels23
Sahni et al., 2020 [88]12Pre-operativeTo identify potential prognostic biomarkers in plasma and isolated microparticles from PDAC patients.NoYesNo—compSWATH-MSPTPRB
PTPRM
Wu et al., 2021 [90]71Stage I–IVTo identify novel plasma glycobiomarkers of pancreatic cancer, with a view to analysing the glycoproteome of plasma samples from patients with non-metastatic and metastatic cancer and gallstones.Yes
(NR, NR)
YesNo—comp (glycoproteome)LC-MS22
Wu et al., 2019 [83]183Stage I–IVTo identify proteomic changes in sera of pancreatic cancer patients, and subsequently evaluate the expression levels of these proteins to evaluate their potential as possible diagnostic markers.Yes
[0.966]
NoNo—compLC-MSPROZ
TNFRSF6B
Yu et al., 2021 [86]135Stage I–IVTo identify plasma protein biomarkers for early detection of PDAC.Yes
[0.81–0.85]
(2 cohorts) b
NoYesOlink—oncology II panel8
Notes: N denotes number of PDAC patients included in study only. Most appropriate descriptor of disease has been applied, including AJCC (8th) disease stage or resection state. Diagnostic performance is reported as AUROC where possible, or sensitivity and specificity; reported relative to healthy controls, without stratification by disease stage, and without combination with other metrics (e.g., CA19-9). Protein(s) examined refers to those explored in detail, and is presented as names if ≤5 reported, or number of proteins if >5 reported. All numbers reported to three significant figures, wherever possible. Abbreviations: AUROC, area under receiver operator curve; NR, not reported (or unable to be deconvoluted from reported data); comp, comprehensive; MS, mass spectrometry; DM-LCT-MS, dimethylation liquid chromatography tandem MS; LC-ESI-MS, liquid chromatography electrospray ionisation MS; UPLC-UDMS, ultra-performance liquid chromatography ultra-definition MS; MRM-MS, multiple reaction monitoring MS; SWATH-MS, sequential window acquisition of all theoretical mass spectra; LC-MS, liquid chromatography MS. a Diagnostic utility reported as significant improvement in C-statistic rather than AUROC or sensitivity/specificity. b AUROC reported as range of published values for two independent cohorts.
Table 5. Summary of relative limitations and strengths of biomarker modalities.
Table 5. Summary of relative limitations and strengths of biomarker modalities.
ModalityLimitationsStrengths
ctDNA
Low diagnostic test sensitivity, especially in early disease;
Limited studies exploring tumour-informed approaches.
Good evidence for prognostication and predictive therapeutic response monitoring.
CTCs
Low diagnostic test sensitivity, especially in early disease;
Prognostic study discrepancies between published works;
Analysis methods display intrinsic inaccuracies;
Rarity and low CTC yield from patient blood samples.
Highly specific (CTCs absent in healthy individuals);
Evidence suggests sub-analysis of CTC subpopulations may improve test performance.
Exosomes
Relative paucity of published papers to date;
Low sample size for many recent explorations;
Lack of validation and corroboration between authors.
Good test performance for diagnosis and prognosis.
Proteomics
Relative paucity of published papers to date;
Prognostication not yet comprehensively explored;
Lack of validation and corroboration between authors;
Several studies to date explored only candidate proteins.
Great diagnostic test performance, especially in conjunction with CA19-9; currently boasts highest reported AUROC values.
Abbreviations: ctDNA, circulating tumour DNA; CTCs, circulating tumour cells.

3.5. Multi-Cancer Early Detection Tests

Beyond explorations of PDAC in isolation, recent studies have also investigated multi-cancer early detection (MCED) tests that utilise liquid biopsy techniques. These assays seek to detect early-stage tumours for multiple cancer types, with a view for implementation as a population screening tool. Notable MCETs to date include CancerSEEK and Galleri.
CancerSEEK is a MCED test developed by Cohen et al. [92] able to detect eight common solid cancers using patient plasma, including pancreatic cancer. The test utilises a combination of ctDNA mutational analysis, involving multiplex PCR-based analysis of sixteen genes (61 amplicons), and measurement of eight circulating proteins via immunoassay. The study examined 1005 patients diagnosed with stage I-III non-metastatic tumours, including 93 patients with pancreatic cancer. Diagnostic sensitivity varied by cancer type from 69 to 98%, with an overall sensitivity for pancreatic cancer detection of 83.7% (and 99.5% specificity) [92]. Diagnostic performance improved with disease stage, with a reported test sensitivity of approximately 60% for stage I–II disease [93].
Conversely, Galleri is a MCED test developed by biotechnology company GRAIL based on data obtained from the Circulating Cell-free Genome Atlas (CCGA) study [94]. The test utilises a targeted ctDNA methylation assay combined with machine learning algorithms for population cancer screening [95]. The most recent substudy involved an analysis of 2823 patients across 27 types of cancer, including pancreatic cancer, spanning stages I-IV. Overall test sensitivity and specificity were 51.5% and 99.5%, respectively, with a higher-than-average sensitivity of 83.7% for pancreatic cancer. Like CancerSEEK, sensitivity generally increased with stage, reporting 61.9% sensitivity for stage I pancreatic cancer [94].
An additional two large, prospective, multi-centre, observational cohort studies are currently being conducted to clinically validate Galleri in intended-use populations [96]. STRIVE is a GRAIL study aiming to recruit 120,000 women undergoing screening mammography and is expected to be completed in 2025 [97], whilst SUMMIT is another GRAIL study intending to enrol 25,000 individuals at high risk for lung cancer and is expected to conclude in 2030 [98]. Furthermore, the PATHFINDER study was recently conducted to assess the feasibility of MCED testing for cancer screening by evaluating the impact of positive results on subsequent testing required for diagnostic resolution [96]. Schrag et al. examined 6662 participants prospectively, concluding overall that the study supported the viability of MCED screening; however, a positive predictive value (PPV) of just 38.0% was reported [99].
Therefore, whilst MCED testing may play a vital role in the ongoing development of crucial population screening tools for many cancers, including pancreatic cancer, it is evident further work is needed to achieve optimal clinical utility. Indeed, higher diagnostic sensitivities and improvements in PPV should be prioritised with future iterations of such technology.

4. Conclusions

The rising incidence, late detection, and lethality of PDAC presents an important health issue that necessitates the development of novel diagnostic, prognostic, and predictive biomarkers. Liquid biopsy techniques, including ctDNA, CTCs, circulating tumor exosomes, and proteomics, have emerged as potential solutions, offering the promise of transforming PDAC management and improving survival. Among these, proteomics stands out as a particularly promising avenue for further development, as new research highlights its superior diagnostic accuracy, as well as its potential prognostic value. However, further investigation is needed to validate existing findings, and comprehensively explore the proteome. Additionally, ongoing improvements to MCED testing performance are warranted for future development of effective population cancer screening tools.

Author Contributions

Conceptualisation, R.M. and B.L.; writing—original draft preparation, R.M.; writing—review and editing, R.M., P.G. and B.L.; supervision, B.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

Appendix A

Figure A1. Ovid MEDLINE Search Strategy. Formatting consistent with Ovid syntax. Created with BioRender.
Figure A1. Ovid MEDLINE Search Strategy. Formatting consistent with Ovid syntax. Created with BioRender.
Cancers 16 03335 g0a1

References

  1. Hu, J.X.; Zhao, C.F.; Chen, W.B.; Liu, Q.C.; Li, Q.W.; Lin, Y.Y.; Gao, F. Pancreatic cancer: A review of epidemiology, trend, and risk factors. World J. Gastroenterol. 2021, 27, 4298–4321. [Google Scholar] [CrossRef] [PubMed]
  2. Kleeff, J.; Korc, M.; Apte, M.; La Vecchia, C.; Johnson, C.D.; Biankin, A.V.; Neale, R.E.; Tempero, M.; Tuveson, D.A.; Hruban, R.H.; et al. Pancreatic cancer. Nat. Rev. Dis. Primers 2016, 2, 16022. [Google Scholar] [CrossRef] [PubMed]
  3. Sarantis, P.; Koustas, E.; Papadimitropoulou, A.; Papavassiliou, A.G.; Karamouzis, M.V. Pancreatic ductal adenocarcinoma: Treatment hurdles, tumor microenvironment and immunotherapy. World J. Gastrointest. Oncol. 2020, 12, 173–181. [Google Scholar] [CrossRef] [PubMed]
  4. AIHW. Cancer Data in Australia; AIHW: Canberra, Australia, 2022. [Google Scholar]
  5. Meng, R.; Chen, J.W.; D’Onise, K.; Barreto, S.G. Pancreatic ductal adenocarcinoma survival in South Australia: Time trends and impact of tumour location. ANZ J. Surg. 2021, 91, 921–926. [Google Scholar] [CrossRef] [PubMed]
  6. Ushio, J.; Kanno, A.; Ikeda, E.; Ando, K.; Nagai, H.; Miwata, T.; Kawasaki, Y.; Tada, Y.; Yokoyama, K.; Numao, N.; et al. Pancreatic Ductal Adenocarcinoma: Epidemiology and Risk Factors. Diagnostics 2021, 11, 562. [Google Scholar] [CrossRef] [PubMed]
  7. Jiang, Y.; Sohal, D.P.S. Pancreatic Adenocarcinoma Management. JCO Oncol. Pract. 2022, 19, 19–32. [Google Scholar] [CrossRef]
  8. Nakaoka, K.; Ohno, E.; Kawabe, N.; Kuzuya, T.; Funasaka, K.; Nakagawa, Y.; Nagasaka, M.; Ishikawa, T.; Watanabe, A.; Tochio, T.; et al. Current Status of the Diagnosis of Early-Stage Pancreatic Ductal Adenocarcinoma. Diagnostics 2023, 13, 215. [Google Scholar] [CrossRef]
  9. Takikawa, T.; Kikuta, K.; Hamada, S.; Kume, K.; Miura, S.; Yoshida, N.; Tanaka, Y.; Matsumoto, R.; Ikeda, M.; Kataoka, F.; et al. Clinical features and prognostic impact of asymptomatic pancreatic cancer. Sci. Rep. 2022, 12, 4262. [Google Scholar] [CrossRef] [PubMed]
  10. Tempero, M.A.; Malafa, M.P.; Al-Hawary, M.; Asbun, H.; Bain, A.; Behrman, S.W.; Benson, A.B., III; Binder, E.; Cardin, D.B.; Cha, C.; et al. Pancreatic Adenocarcinoma, Version 2.2017, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2017, 15, 1028–1061. [Google Scholar] [CrossRef]
  11. Sharma, A.; Kandlakunta, H.; Nagpal, S.J.S.; Feng, Z.; Hoos, W.; Petersen, G.M.; Chari, S.T. Model to Determine Risk of Pancreatic Cancer in Patients with New-Onset Diabetes. Gastroenterology 2018, 155, 730–739.e733. [Google Scholar] [CrossRef] [PubMed]
  12. Tiwari, A.; Kumar, L. Pancreatic ductal adenocarcinoma: Role of chemotherapy & future perspectives. Indian J. Med. Res. 2018, 148, 254–257. [Google Scholar] [CrossRef] [PubMed]
  13. Yang, H.; Li, W.; Ren, L.; Yang, Y.; Zhang, Y.; Ge, B.; Li, S.; Zheng, X.; Liu, J.; Zhang, S.; et al. Progress on diagnostic and prognostic markers of pancreatic cancer. Oncol. Res. 2023, 31, 83–99. [Google Scholar] [CrossRef] [PubMed]
  14. Ballehaninna, U.K.; Chamberlain, R.S. The clinical utility of serum CA 19-9 in the diagnosis, prognosis and management of pancreatic adenocarcinoma: An evidence based appraisal. J. Gastrointest. Oncol. 2012, 3, 105–119. [Google Scholar] [CrossRef] [PubMed]
  15. Luo, G.; Jin, K.; Deng, S.; Cheng, H.; Fan, Z.; Gong, Y.; Qian, Y.; Huang, Q.; Ni, Q.; Liu, C.; et al. Roles of CA19-9 in pancreatic cancer: Biomarker, predictor and promoter. Biochim. Biophys. Acta BBA Rev. Cancer 2021, 1875, 188409. [Google Scholar] [CrossRef]
  16. Hartwig, W.; Strobel, O.; Hinz, U.; Fritz, S.; Hackert, T.; Roth, C.; Büchler, M.W.; Werner, J. CA19-9 in Potentially Resectable Pancreatic Cancer: Perspective to Adjust Surgical and Perioperative Therapy. Ann. Surg. Oncol. 2013, 20, 2188–2196. [Google Scholar] [CrossRef] [PubMed]
  17. Isaji, S.; Mizuno, S.; Windsor, J.A.; Bassi, C.; Fernández-Del Castillo, C.; Hackert, T.; Hayasaki, A.; Katz, M.H.G.; Kim, S.W.; Kishiwada, M.; et al. International consensus on definition and criteria of borderline resectable pancreatic ductal adenocarcinoma 2017. Pancreatology 2018, 18, 2–11. [Google Scholar] [CrossRef]
  18. Kang, M.J.; Kim, S.-W. Paradigm shandt for defining the resectability of pancreatic cancer. Ann. Hepatobiliary Pancreat. Surg. 2021, 25, 451–455. [Google Scholar] [CrossRef] [PubMed]
  19. Nong, M.Z.; Dove, D.; Fischer, D.A.; Hourdequin, K.C.; Ripple, G.H.; Amin, M.A.; McGrath, E.B.; Zaki, B.I.; Smith, K.D.; Brooks, G.A. Surveillance with Serial Imaging and CA 19-9 Tumor Marker Testing after Resection of Pancreatic Cancer: A Single-Center Retrospective Study. Am. J. Clin. Oncol. 2024, 47, 25–29. [Google Scholar] [CrossRef]
  20. Lee, T.; Teng, T.Z.J.; Shelat, V.G. Carbohydrate antigen 19-9—Tumor marker: Past, present, and future. World J. Gastrointest. Surg. 2020, 12, 468–490. [Google Scholar] [CrossRef]
  21. Watanabe, F.; Suzuki, K.; Noda, H.; Rikiyama, T. Liquid biopsy leads to a paradigm shift in the treatment of pancreatic cancer. World J. Gastroenterol. 2022, 28, 6478–6496. [Google Scholar] [CrossRef]
  22. Sherman, M.H.; Beatty, G.L. Tumor Microenvironment in Pancreatic Cancer Pathogenesis and Therapeutic Resistance. Ann. Rev. Pathol. 2023, 18, 123–148. [Google Scholar] [CrossRef] [PubMed]
  23. Elhanafi, S.; Mahmud, N.; Vergara, N.; Kochman, M.L.; Das, K.K.; Ginsberg, G.G.; Rajala, M.; Chandrasekhara, V. Comparison of endoscopic ultrasound tissue acquisition methods for genomic analysis of pancreatic cancer. J. Gastroenterol. Hepatol. 2019, 34, 907–913. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, K.; Wang, X.; Pan, Q.; Zhao, B. Liquid biopsy techniques and pancreatic cancer: Diagnosis, monitoring, and evaluation. Mol. Cancer 2023, 22, 167. [Google Scholar] [CrossRef] [PubMed]
  25. Chudasama, D.; Katopodis, P.; Stone, N.; Haskell, J.; Sheridan, H.; Gardner, B.; Urnovitz, H.; Schuetz, E.; Beck, J.; Hall, M.; et al. Liquid Biopsies in Lung Cancer: Four Emerging Technologies and Potential Clinical Applications. Cancers 2019, 11, 331. [Google Scholar] [CrossRef]
  26. Stefanovic, S.; Deutsch, T.M.; Wirtz, R.; Hartkopf, A.; Sinn, P.; Schuetz, F.; Sohn, C.; Bohlmann, M.K.; Sütterlin, M.; Schneeweiss, A.; et al. Molecular Subtype Conversion between Primary and Metastatic Breast Cancer Corresponding to the Dynamics of Apoptotic and Intact Circulating Tumor Cells. Cancers 2019, 11, 342. [Google Scholar] [CrossRef] [PubMed]
  27. Lee, B.; Tie, J.; Wang, Y.; Cohen, J.D.; Shapiro, J.D.; Wong, R.; Aghmesheh, M.; Kiberu, A.D.; Francesconi, A.; Burge, M.E.; et al. The potential role of serial circulating tumor DNA (ctDNA) testing after upfront surgery to guide adjuvant chemotherapy for early stage pancreatic cancer: The AGITG DYNAMIC-Pancreas trial. J. Clin. Oncol. 2024, 42, 107. [Google Scholar] [CrossRef]
  28. Lee, J.; Ahn, J.; Jung, K.; Lee, J.-c.; Youn, Y.; Hwang, J.-H. Serial circulating tumor DNA monitoring for predicting treatment response and prognosis in patients with pancreatic cancer. J. Clin. Oncol. 2024, 42, 4154. [Google Scholar] [CrossRef]
  29. Buscail, E.; Maulat, C.; Muscari, F.; Chiche, L.; Cordelier, P.; Dabernat, S.; Alix-Panabières, C.; Buscail, L. Liquid Biopsy Approach for Pancreatic Ductal Adenocarcinoma. Cancers 2019, 11, 852. [Google Scholar] [CrossRef]
  30. Bronkhorst, A.J.; Ungerer, V.; Holdenrieder, S. The emerging role of cell-free DNA as a molecular marker for cancer management. Biomol. Detect. Quantif. 2019, 17, 100087. [Google Scholar] [CrossRef] [PubMed]
  31. Topham, J.T.; Renouf, D.J.; Schaeffer, D.F. Circulating tumor DNA: Toward evolving the clinical paradigm of pancreatic ductal adenocarcinoma. Ther. Adv. Med. Oncol. 2023, 15, 17588359231157651. [Google Scholar] [CrossRef]
  32. Cohen, R.; Platell, C.F.; McCoy, M.J.; Meehan, K.; Fuller, K. Circulating tumour DNA in colorectal cancer management. Br. J. Surg. 2023, 110, 773–783. [Google Scholar] [CrossRef] [PubMed]
  33. Honoré, N.; van Marcke, C.; Galot, R.; Helaers, R.; Ambroise, J.; van Maanen, A.; Mendola, A.; Dahou, H.; Marbaix, E.; Van Eeckhout, P.; et al. Tumor-agnostic plasma assay for circulating tumor DNA detects minimal residual disease and predicts outcome in locally advanced squamous cell carcinoma of the head and neck. Ann. Oncol. 2023, 34, 1175–1186. [Google Scholar] [CrossRef] [PubMed]
  34. Chan, H.T.; Nagayama, S.; Otaki, M.; Chin, Y.M.; Fukunaga, Y.; Ueno, M.; Nakamura, Y.; Low, S.K. Tumor-informed or tumor-agnostic circulating tumor DNA as a biomarker for risk of recurrence in resected colorectal cancer patients. Front. Oncol. 2022, 12, 1055968. [Google Scholar] [CrossRef] [PubMed]
  35. Huerta, M.; Roselló, S.; Sabater, L.; Ferrer, A.; Tarazona, N.; Roda, D.; Gambardella, V.; Alfaro-Cervelló, C.; Garcés-Albir, M.; Cervantes, A.; et al. Circulating Tumor DNA Detection by Digital-Droplet PCR in Pancreatic Ductal Adenocarcinoma: A Systematic Review. Cancers 2021, 13, 994. [Google Scholar] [CrossRef]
  36. Macgregor-Das, A.; Yu, J.; Tamura, K.; Abe, T.; Suenaga, M.; Shindo, K.; Borges, M.; Koi, C.; Kohi, S.; Sadakari, Y.; et al. Detection of Circulating Tumor DNA in Patients with Pancreatic Cancer Using Digital Next-Generation Sequencing. J. Mol. Diagn. JMD 2020, 22, 748–756. [Google Scholar] [CrossRef] [PubMed]
  37. Hussung, S.; Akhoundova, D.; Hipp, J.; Follo, M.; Klar, R.F.U.; Philipp, U.; Scherer, F.; von Bubnoff, N.; Duyster, J.; Boerries, M.; et al. Longitudinal analysis of cell-free mutated KRAS and CA 19-9 predicts survival following curative resection of pancreatic cancer. BMC Cancer 2021, 21, 49. [Google Scholar] [CrossRef] [PubMed]
  38. Terasawa, H.; Kinugasa, H.; Ako, S.; Hirai, M.; Matsushita, H.; Uchida, D.; Tomoda, T.; Matsumoto, K.; Horiguchi, S.; Kato, H.; et al. Utility of liquid biopsy using urine in patients with pancreatic ductal adenocarcinoma. Cancer Biol. Ther. 2019, 20, 1348–1353. [Google Scholar] [CrossRef] [PubMed]
  39. Groot, V.P.; Mosier, S.; Javed, A.A.; Teinor, J.A.; Gemenetzis, G.; Ding, D.; Haley, L.M.; Yu, J.; Burkhart, R.A.; Hasanain, A.; et al. Circulating Tumor DNA as a Clinical Test in Resected Pancreatic Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 4973–4984. [Google Scholar] [CrossRef]
  40. Lee, B.; Lipton, L.; Cohen, J.; Tie, J.; Javed, A.A.; Li, L.; Goldstein, D.; Burge, M.; Cooray, P.; Nagrial, A.; et al. Circulating tumor DNA as a potential marker of adjuvant chemotherapy benefit following surgery for localized pancreatic cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2019, 30, 1472–1478. [Google Scholar] [CrossRef]
  41. Guo, S.; Shi, X.; Shen, J.; Gao, S.; Wang, H.; Shen, S.; Pan, Y.; Li, B.; Xu, X.; Shao, Z.; et al. Preoperative detection of KRAS G12D mutation in ctDNA is a powerful predictor for early recurrence of resectable PDAC patients. Br. J. Cancer 2020, 122, 857–867. [Google Scholar] [CrossRef]
  42. Chung, C.; Galvin, R.; Achenbach, E.; Dziadkowiec, O.; Sen, S. Characterization of Blood-Based Molecular Profiling in Pancreatic Adenocarcinoma. Oncology 2021, 35, 794–803. [Google Scholar] [CrossRef] [PubMed]
  43. Strijker, M.; Soer, E.C.; de Pastena, M.; Creemers, A.; Balduzzi, A.; Beagan, J.J.; Busch, O.R.; van Delden, O.M.; Halfwerk, H.; van Hooft, J.E.; et al. Circulating tumor DNA quantity is related to tumor volume and both predict survival in metastatic pancreatic ductal adenocarcinoma. Int. J. Cancer 2020, 146, 1445–1456. [Google Scholar] [CrossRef] [PubMed]
  44. Uesato, Y.; Sasahira, N.; Ozaka, M.; Sasaki, T.; Takatsuki, M.; Zembutsu, H. Evaluation of circulating tumor DNA as a biomarker in pancreatic cancer with liver metastasis. PLoS ONE 2020, 15, e0235623. [Google Scholar] [CrossRef] [PubMed]
  45. Wei, T.; Zhang, Q.; Li, X.; Su, W.; Li, G.; Ma, T.; Gao, S.; Lou, J.; Que, R.; Zheng, L.; et al. Monitoring Tumor Burden in Response to FOLFIRINOX Chemotherapy via Profiling Circulating Cell-Free DNA in Pancreatic Cancer. Mol. Cancer Ther. 2019, 18, 196–203. [Google Scholar] [CrossRef]
  46. Bachet, J.-B.; Blons, H.; Hammel, P.; Hariry, I.E.; Portales, F.; Mineur, L.; Metges, J.-P.; Mulot, C.; Bourreau, C.; Cain, J.; et al. Circulating Tumor DNA is Prognostic and Potentially Predictive of Eryaspase Efficacy in Second-line in Patients with Advanced Pancreatic Adenocarcinoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 5208–5216. [Google Scholar] [CrossRef]
  47. Patel, H.; Okamura, R.; Fanta, P.; Patel, C.; Lanman, R.B.; Raymond, V.M.; Kato, S.; Kurzrock, R. Clinical correlates of blood-derived circulating tumor DNA in pancreatic cancer. J. Hematol. Oncol. 2019, 12, 130. [Google Scholar] [CrossRef]
  48. Kim, M.P.; Li, X.; Deng, J.; Zhang, Y.; Dai, B.; Allton, K.L.; Hughes, T.G.; Siangco, C.; Augustine, J.J.; Kang, Y.; et al. Oncogenic KRAS Recruits an Expansive Transcriptional Network through Mutant p53 to Drive Pancreatic Cancer Metastasis. Cancer Discov. 2021, 11, 2094–2111. [Google Scholar] [CrossRef]
  49. Sivapalan, L.; Kocher, H.M.; Ross-Adams, H.; Chelala, C. Molecular profiling of ctDNA in pancreatic cancer: Opportunities and challenges for clinical application. Pancreatology 2021, 21, 363–378. [Google Scholar] [CrossRef]
  50. Alese, O.B.; Cook, N.; Ortega-Franco, A.; Ulanja, M.B.; Tan, L.; Tie, J. Circulating Tumor DNA: An Emerging Tool in Gastrointestinal Cancers. Am. Soc. Clin. Oncol. Educ. Book 2022, 40, 279–298. [Google Scholar] [CrossRef]
  51. Pietrasz, D.; Wang-Renault, S.; Taieb, J.; Dahan, L.; Postel, M.; Durand-Labrunie, J.; Le Malicot, K.; Mulot, C.; Rinaldi, Y.; Phelip, J.-M.; et al. Prognostic value of circulating tumour DNA in metastatic pancreatic cancer patients: Post-hoc analyses of two clinical trials. Br. J. Cancer 2022, 126, 440–448. [Google Scholar] [CrossRef]
  52. Singh, N.; Rashid, S.; Rashid, S.; Dash, N.R.; Gupta, S.; Saraya, A. Clinical significance of promoter methylation status of tumor suppressor genes in circulating DNA of pancreatic cancer patients. J. Cancer Res. Clin. Oncol. 2020, 146, 897–907. [Google Scholar] [CrossRef] [PubMed]
  53. Miller, B.F.; Petrykowska, H.M.; Elnitski, L. Assessing ZNF154 methylation in patient plasma as a multicancer marker in liquid biopsies from colon, liver, ovarian and pancreatic cancer patients. Sci. Rep. 2021, 11, 221. [Google Scholar] [CrossRef] [PubMed]
  54. Shinjo, K.; Hara, K.; Nagae, G.; Umeda, T.; Katsushima, K.; Suzuki, M.; Murofushi, Y.; Umezu, Y.; Takeuchi, I.; Takahashi, S.; et al. A novel sensitive detection method for DNA methylation in circulating free DNA of pancreatic cancer. PLoS ONE 2020, 15, e0233782. [Google Scholar] [CrossRef]
  55. Fujimoto, Y.; Suehiro, Y.; Kaino, S.; Suenaga, S.; Tsuyama, T.; Matsui, H.; Higaki, S.; Fujii, I.; Suzuki, C.; Hoshida, T.; et al. Combination of CA19-9 and Blood Free-Circulating Methylated RUNX3 May Be Useful to Diagnose Stage I Pancreatic Cancer. Oncology 2021, 99, 234–239. [Google Scholar] [CrossRef] [PubMed]
  56. Li, S.; Zhang, G.; Li, X.; Li, X.; Chen, X.; Xu, Y.; Ren, H. Role of the preoperative circulating tumor DNA KRAS mutation in patients with resectable pancreatic cancer. Pharmacogenomics 2021, 22, 657–667. [Google Scholar] [CrossRef]
  57. Sugimori, M.; Sugimori, K.; Tsuchiya, H.; Suzuki, Y.; Tsuyuki, S.; Kaneta, Y.; Hirotani, A.; Sanga, K.; Tozuka, Y.; Komiyama, S.; et al. Quantitative monitoring of circulating tumor DNA in patients with advanced pancreatic cancer undergoing chemotherapy. Cancer Sci. 2020, 111, 266–278. [Google Scholar] [CrossRef]
  58. Cheng, H.; Luo, G.; Jin, K.; Fan, Z.; Huang, Q.; Gong, Y.; Xu, J.; Yu, X.; Liu, C. Kras mutation correlating with circulating regulatory T cells predicts the prognosis of advanced pancreatic cancer patients. Cancer Med. 2020, 9, 2153–2159. [Google Scholar] [CrossRef]
  59. Watanabe, F.; Suzuki, K.; Tamaki, S.; Abe, I.; Endo, Y.; Takayama, Y.; Ishikawa, H.; Kakizawa, N.; Saito, M.; Futsuhara, K.; et al. Longitudinal monitoring of KRAS-mutated circulating tumor DNA enables the prediction of prognosis and therapeutic responses in patients with pancreatic cancer. PLoS ONE 2019, 14, e0227366. [Google Scholar] [CrossRef] [PubMed]
  60. Wang, S.-E.; Shyr, B.-U.; Shyr, B.-S.; Chen, S.-C.; Chang, S.-C.; Shyr, Y.-M. Circulating Cell-Free DNA in Pancreatic Head Adenocarcinoma Undergoing Pancreaticoduodenectomy. Pancreas 2021, 50, 214–218. [Google Scholar] [CrossRef]
  61. Thiele, J.A.; Pitule, P.; Hicks, J.; Kuhn, P. Single-Cell Analysis of Circulating Tumor Cells. Methods Mol. Biol. 2019, 1908, 243–264. [Google Scholar] [CrossRef]
  62. Xing, Y.; Zhang, X.; Qin, F.; Yang, J.; Ai, L.; Wang, Q.; Zhai, Y. The clinical significance of circulating tumor cells and T lymphocyte subtypes in pancreatic cancer patients. Bioengineered 2022, 13, 2130–2138. [Google Scholar] [CrossRef] [PubMed]
  63. Zhu, P.; Liu, H.-Y.; Liu, F.-C.; Gu, F.-M.; Yuan, S.-X.; Huang, J.; Pan, Z.-Y.; Wang, W.-J. Circulating Tumor Cells Expressing Kruppel-Like Factor 8 and Vimentin as Predictors of Poor Prognosis in Pancreatic Cancer Patients. Cancer Control J. Moffitt Cancer Cent. 2021, 28, 10732748211027163. [Google Scholar] [CrossRef] [PubMed]
  64. Wei, T.; Zhang, X.; Zhang, Q.; Yang, J.; Chen, Q.; Wang, J.; Li, X.; Chen, J.; Ma, T.; Li, G.; et al. Vimentin-positive circulating tumor cells as a biomarker for diagnosis and treatment monitoring in patients with pancreatic cancer. Cancer Lett. 2019, 452, 237–243. [Google Scholar] [CrossRef] [PubMed]
  65. Semaan, A.; Bernard, V.; Kim, D.U.; Lee, J.J.; Huang, J.; Kamyabi, N.; Stephens, B.M.; Qiao, W.; Varadhachary, G.R.; Katz, M.H.; et al. Characterisation of circulating tumour cell phenotypes identifies a partial-EMT sub-population for clinical stratification of pancreatic cancer. Br. J. Cancer 2021, 124, 1970–1977. [Google Scholar] [CrossRef] [PubMed]
  66. Zhao, X.-H.; Wang, Z.-R.; Chen, C.-L.; Di, L.; Bi, Z.-F.; Li, Z.-H.; Liu, Y.-M. Molecular detection of epithelial-mesenchymal transition markers in circulating tumor cells from pancreatic cancer patients: Potential role in clinical practice. World J. Gastroenterol. 2019, 25, 138–150. [Google Scholar] [CrossRef] [PubMed]
  67. Park, Y.; Jun, H.R.; Choi, H.W.; Hwang, D.W.; Lee, J.H.; Song, K.B.; Lee, W.; Kwon, J.; Ha, S.H.; Jun, E.; et al. Circulating tumour cells as an indicator of early and systemic recurrence after surgical resection in pancreatic ductal adenocarcinoma. Sci. Rep. 2021, 11, 1644. [Google Scholar] [CrossRef]
  68. Cheng, H.; He, W.; Yang, J.; Ye, Q.; Cheng, L.; Pan, Y.; Mao, L.; Chu, X.; Lu, C.; Li, G.; et al. Ligand-targeted polymerase chain reaction for the detection of folate receptor-positive circulating tumour cells as a potential diagnostic biomarker for pancreatic cancer. Cell Prolif. 2020, 53, e12880. [Google Scholar] [CrossRef]
  69. Aiello, N.M.; Kang, Y. Context-dependent EMT programs in cancer metastasis. J. Exp. Med. 2019, 216, 1016–1026. [Google Scholar] [CrossRef] [PubMed]
  70. Gasparini-Junior, J.L.; Fanelli, M.F.; Abdallah, E.A.; Chinen, L.T.D. Evaluating Mmp-2 and Tgfs-Ri Expression in Circulating Tumor Cells Of Pancreatic Cancer Patients and Their Correlation with Clinical Evolution. Arq. Bras. Cir. Dig. ABCD Braz. Arch. Dig. Surg. 2019, 32, e1433. [Google Scholar] [CrossRef]
  71. Stoecklein, N.H.; Fluegen, G.; Guglielmi, R.; Neves, R.P.L.; Hackert, T.; Birgin, E.; Cieslik, S.A.; Sudarsanam, M.; Driemel, C.; van Dalum, G.; et al. Ultra-sensitive CTC-based liquid biopsy for pancreatic cancer enabled by large blood volume analysis. Mol. Cancer 2023, 22, 181. [Google Scholar] [CrossRef]
  72. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef] [PubMed]
  73. Takahashi, K.; Ota, Y.; Kogure, T.; Suzuki, Y.; Iwamoto, H.; Yamakita, K.; Kitano, Y.; Fujii, S.; Haneda, M.; Patel, T.; et al. Circulating extracellular vesicle-encapsulated HULC is a potential biomarker for human pancreatic cancer. Cancer Sci. 2020, 111, 98–111. [Google Scholar] [CrossRef] [PubMed]
  74. Kitagawa, T.; Taniuchi, K.; Tsuboi, M.; Sakaguchi, M.; Kohsaki, T.; Okabayashi, T.; Saibara, T. Circulating pancreatic cancer exosomal RNAs for detection of pancreatic cancer. Mol. Oncol. 2019, 13, 212–227. [Google Scholar] [CrossRef]
  75. Vicentini, C.; Calore, F.; Nigita, G.; Fadda, P.; Simbolo, M.; Sperandio, N.; Luchini, C.; Lawlor, R.T.; Croce, C.M.; Corbo, V.; et al. Exosomal miRNA signatures of pancreatic lesions. BMC Gastroenterol. 2020, 20, 137. [Google Scholar] [CrossRef] [PubMed]
  76. Tao, L.; Zhou, J.; Yuan, C.; Zhang, L.; Li, D.; Si, D.; Xiu, D.; Zhong, L. Metabolomics identifies serum and exosomes metabolite markers of pancreatic cancer. Metabolomics Off. J. Metabolomic Soc. 2019, 15, 86. [Google Scholar] [CrossRef]
  77. Wei, Q.; Wei, L.; Zhang, J.; Li, Z.; Feng, H.; Ren, L. EphA2-enriched exosomes promote cell migration and are a potential diagnostic serum marker in pancreatic cancer. Mol. Med. Rep. 2020, 22, 2941–2947. [Google Scholar] [CrossRef] [PubMed]
  78. Wei, Q.; Zhang, J.; Li, Z.; Wei, L.; Ren, L. Serum Exo-EphA2 as a Potential Diagnostic Biomarker for Pancreatic Cancer. Pancreas 2020, 49, 1213–1219. [Google Scholar] [CrossRef]
  79. Xiao, D.; Dong, Z.; Zhen, L.; Xia, G.; Huang, X.; Wang, T.; Guo, H.; Yang, B.; Xu, C.; Wu, W.; et al. Combined Exosomal GPC1, CD82, and Serum CA19-9 as Multiplex Targets: A Specific, Sensitive, and Reproducible Detection Panel for the Diagnosis of Pancreatic Cancer. Mol. Cancer Res. MCR 2020, 18, 300–310. [Google Scholar] [CrossRef]
  80. Lux, A.; Kahlert, C.; Grutzmann, R.; Pilarsky, C. c-Met and PD-L1 on Circulating Exosomes as Diagnostic and Prognostic Markers for Pancreatic Cancer. Int. J. Mol. Sci. 2019, 20, 3305. [Google Scholar] [CrossRef] [PubMed]
  81. Giampieri, R.; Piva, F.; Occhipinti, G.; Bittoni, A.; Righetti, A.; Pagliaretta, S.; Murrone, A.; Bianchi, F.; Amantini, C.; Giulietti, M.; et al. Clinical impact of different exosomes’ protein expression in pancreatic ductal carcinoma patients treated with standard first line palliative chemotherapy. PLoS ONE 2019, 14, e0215990. [Google Scholar] [CrossRef] [PubMed]
  82. Kim, Y.; Yeo, I.; Huh, I.; Kim, J.; Han, D.; Jang, J.-Y.; Kim, Y. Development and Multiple Validation of the Protein Multi-marker Panel for Diagnosis of Pancreatic Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 2236–2245. [Google Scholar] [CrossRef] [PubMed]
  83. Wu, X.; Zhang, Z.-X.; Chen, X.-Y.; Xu, Y.-L.; Yin, N.; Yang, J.; Zhu, D.-M.; Li, D.-C.; Zhou, J. A Panel of Three Biomarkers Identified by iTRAQ for the Early Diagnosis of Pancreatic Cancer. Proteom. Clin. Appl. 2019, 13, e1800195. [Google Scholar] [CrossRef] [PubMed]
  84. Kartsonaki, C.; Pang, Y.; Millwood, I.; Yang, L.; Guo, Y.; Walters, R.; Lv, J.; Hill, M.; Yu, C.; Chen, Y.; et al. Circulating proteins and risk of pancreatic cancer: A case-subcohort study among Chinese adults. Int. J. Epidemiol. 2022, 51, 817–829. [Google Scholar] [CrossRef]
  85. Duan, B.; Hu, X.; Fan, M.; Xiong, X.; Han, L.; Wang, Z.; Tong, D.; Liu, L.; Wang, X.; Li, W.; et al. RNA-Binding Motif Protein 6 is a Candidate Serum Biomarker for Pancreatic Cancer. Proteom. Clin. Appl. 2019, 13, e1900048. [Google Scholar] [CrossRef] [PubMed]
  86. Yu, J.; Ploner, A.; Kordes, M.; Lohr, M.; Nilsson, M.; de Maturana, M.E.L.; Estudillo, L.; Renz, H.; Carrato, A.; Molero, X.; et al. Plasma protein biomarkers for early detection of pancreatic ductal adenocarcinoma. Int. J. Cancer 2021, 148, 2048–2058. [Google Scholar] [CrossRef] [PubMed]
  87. Deutsch, O.; Haviv, Y.; Krief, G.; Keshet, N.; Westreich, R.; Stemmer, S.M.; Zaks, B.; Navat, S.P.; Yanko, R.; Lahav, O.; et al. Possible proteomic biomarkers for the detection of pancreatic cancer in oral fluids. Sci. Rep. 2020, 10, 21995. [Google Scholar] [CrossRef]
  88. Sahni, S.; Krisp, C.; Molloy, M.P.; Nahm, C.; Maloney, S.; Gillson, J.; Gill, A.J.; Samra, J.; Mittal, A. PSMD11, PTPRM and PTPRB as novel biomarkers of pancreatic cancer progression. Biochim. Biophys. Acta Gen. Subj. 2020, 1864, 129682. [Google Scholar] [CrossRef]
  89. Holm, M.; Saraswat, M.; Joenvaara, S.; Seppanen, H.; Renkonen, R.; Haglund, C. Label-free proteomics reveals serum proteins whose levels differ between pancreatic ductal adenocarcinoma patients with short or long survival. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2020, 42, 1010428320936410. [Google Scholar] [CrossRef]
  90. Wu, C.-C.; Lu, Y.-T.; Yeh, T.-S.; Chan, Y.-H.; Dash, S.; Yu, J.-S. Identification of Fucosylated SERPINA1 as a Novel Plasma Marker for Pancreatic Cancer Using Lectin Affinity Capture Coupled with iTRAQ-Based Quantitative Glycoproteomics. Int. J. Mol. Sci. 2021, 22, 6079. [Google Scholar] [CrossRef]
  91. Rittmann, M.C.; Hussung, S.; Braun, L.M.; Klar, R.F.U.; Biesel, E.A.; Fichtner-Feigl, S.; Fritsch, R.; Wittel, U.A.; Ruess, D.A. Plasma biomarkers for prediction of early tumor recurrence after resection of pancreatic ductal adenocarcinoma. Sci. Rep. 2021, 11, 7499. [Google Scholar] [CrossRef]
  92. Cohen, J.D.; Li, L.; Wang, Y.; Thoburn, C.; Afsari, B.; Danilova, L.; Douville, C.; Javed, A.A.; Wong, F.; Mattox, A.; et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 2018, 359, 926–930. [Google Scholar] [CrossRef] [PubMed]
  93. Nené, N.R.; Ney, A.; Nazarenko, T.; Blyuss, O.; Johnston, H.E.; Whitwell, H.J.; Sedlak, E.; Gentry-Maharaj, A.; Apostolidou, S.; Costello, E.; et al. Serum biomarker-based early detection of pancreatic ductal adenocarcinomas with ensemble learning. Commun. Med. 2023, 3, 10. [Google Scholar] [CrossRef]
  94. Klein, E.A.; Richards, D.; Cohn, A.; Tummala, M.; Lapham, R.; Cosgrove, D.; Chung, G.; Clement, J.; Gao, J.; Hunkapiller, N.; et al. Clinical validation of a targeted methylation-based multi-cancer early detection test using an independent validation set. Ann. Oncol. 2021, 32, 1167–1177. [Google Scholar] [CrossRef]
  95. Liu, M.C.; Oxnard, G.R.; Klein, E.A.; Swanton, C.; Seiden, M.V.; Liu, M.C.; Oxnard, G.R.; Klein, E.A.; Smith, D.; Richards, D.; et al. Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann. Oncol. 2020, 31, 745–759. [Google Scholar] [CrossRef] [PubMed]
  96. Nadauld, L.D.; McDonnell, C.H., III; Beer, T.M.; Liu, M.C.; Klein, E.A.; Hudnut, A.; Whittington, R.A.; Taylor, B.; Oxnard, G.R.; Lipson, J.; et al. The PATHFINDER Study: Assessment of the Implementation of an Investigational Multi-Cancer Early Detection Test into Clinical Practice. Cancers 2021, 13, 3501. [Google Scholar] [CrossRef] [PubMed]
  97. Liu, M.C.; Cummings, S.; Vachon, C.M.; Kerlikowske, K.; Couch, F.J.; Morris, E.A.; Olson, J.E.; Polley, E.C.; Conners, A.L.; Ellis, R.L. Abstract OT3-02-01: Development of cell-free nucleic acid-based tests for early detection of breast cancer: The STRIVE study. Cancer Res. 2018, 78, OT3-02-01–OT03-02-01. [Google Scholar] [CrossRef]
  98. Janes, S.; Dickson, J.; Devaraj, A.; Horst, C.; Quaife, S.; Levermore, C.; Gyertson, K.; Mullin, A.; Farrelly, L.; Allen, B. P1. 11-19 trial in progress: Cancer screening study with or without low dose lung CT to validate a multi-cancer early detection blood test. J. Thorac. Oncol. 2019, 14, S523. [Google Scholar] [CrossRef]
  99. Schrag, D.; Beer, T.M.; McDonnell, C.H., III; Nadauld, L.; Dilaveri, C.A.; Reid, R.; Marinac, C.R.; Chung, K.C.; Lopatin, M.; Fung, E.T.; et al. Blood-based tests for multicancer early detection (PATHFINDER): A prospective cohort study. Lancet 2023, 402, 1251–1260. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PDAC sampling by traditional tissue biopsy and liquid biopsy techniques. Current diagnostic workup for PDAC generally involves initial imaging such as computed tomography or magnetic resonance abdominal imaging, followed by confirmatory histopathological analysis of endoscopic ultrasound-guided fine-needle aspirates. Liquid biopsies present an alternative means to survey tumour-derived biomarkers accessible in peripheral blood, including circulating tumour DNA, circulating tumour cells, circulating tumour exosomes, and circulating free tumour-derived proteins (proteomics). Abbreviations: FNA, fine-needle aspirate; ctDNA, circulating tumour DNA; CTCs, circulating tumour cells. Schematic not to scale. Created with BioRender.
Figure 1. PDAC sampling by traditional tissue biopsy and liquid biopsy techniques. Current diagnostic workup for PDAC generally involves initial imaging such as computed tomography or magnetic resonance abdominal imaging, followed by confirmatory histopathological analysis of endoscopic ultrasound-guided fine-needle aspirates. Liquid biopsies present an alternative means to survey tumour-derived biomarkers accessible in peripheral blood, including circulating tumour DNA, circulating tumour cells, circulating tumour exosomes, and circulating free tumour-derived proteins (proteomics). Abbreviations: FNA, fine-needle aspirate; ctDNA, circulating tumour DNA; CTCs, circulating tumour cells. Schematic not to scale. Created with BioRender.
Cancers 16 03335 g001
Figure 2. PRISMA diagram depicting literature search strategy. The Identification stage initially returned 3147 papers. The Screening and Eligibility stages excluded 2991 papers and 107 papers, respectively. Of the included forty-nine papers, twenty-two pertained to circulating tumour DNA, eight to circulating tumour cells, nine to exosomes, and ten to proteomics.
Figure 2. PRISMA diagram depicting literature search strategy. The Identification stage initially returned 3147 papers. The Screening and Eligibility stages excluded 2991 papers and 107 papers, respectively. Of the included forty-nine papers, twenty-two pertained to circulating tumour DNA, eight to circulating tumour cells, nine to exosomes, and ten to proteomics.
Cancers 16 03335 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Munnings, R.; Gibbs, P.; Lee, B. Evolution of Liquid Biopsies for Detecting Pancreatic Cancer. Cancers 2024, 16, 3335. https://doi.org/10.3390/cancers16193335

AMA Style

Munnings R, Gibbs P, Lee B. Evolution of Liquid Biopsies for Detecting Pancreatic Cancer. Cancers. 2024; 16(19):3335. https://doi.org/10.3390/cancers16193335

Chicago/Turabian Style

Munnings, Ryan, Peter Gibbs, and Belinda Lee. 2024. "Evolution of Liquid Biopsies for Detecting Pancreatic Cancer" Cancers 16, no. 19: 3335. https://doi.org/10.3390/cancers16193335

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop