Background: Pancreatic cancer continues to retain the highest mortality rate among all major organ cancers. New strategies for early detection are being proposed to increase long-term survival. A plethora of molecular markers are discovered yearly, but so far none have demonstrated screening utility. Summary: Promising discovery technologies include affinity-based proteomics and ddPCR. In the validation phase, researchers must decide on key benchmark criteria, what type of pancreatic lesions are desirable to find early through molecular screening, and when to terminate the biomarker study and return to the discovery phase. If the biomarkers meet set benchmarks, retrospective analysis should be conducted in relevant cohorts based on intended use, followed by prospective real-world evaluation. Lastly, regulatory approval, incorporation into clinical practice guidelines, and thorough health economic evaluations must be completed before the screening markers can be fully implemented. Key Messages: In this review, important strategies and phases for molecular biomarker development in pancreatic cancer have been outlined.

In 2024, all major cancers in the EU expected an improved survival trend, except for lung and pancreatic cancer [1]. Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related death, with a majority of cases diagnosed at advanced stages [2]. Acinar-to-ductal metaplasia is implicated in the development of pancreatic intraepithelial neoplasia (PanIN), the most frequent precursor to PDAC. While acinar-to-ductal metaplasia may serve a regenerative function following pancreatic injury or inflammation [3], its persistence can lead to PanIN formation, particularly in the presence of oncogenic KRAS mutations. The progression of PanIN to invasive PDAC is driven by sequential loss of tumor suppressor genes such as p16/CDKN2A, TP53, and SMAD4 [4]. Despite the low incidence of PDAC, PanIN structures in the population exceed 70% in autopsy series [5]. Oncotherapeutic advancements have conferred short-term survival benefits over the past two decades, but the actual chance for long-term survival has remained stagnant since start of systematic data collection [6]. Consequently, more resources have shifted toward the development of biomarker-based screening tools for early detection. This review will focus on current techniques, advances, and important considerations in each area of PDAC early detection, focusing on minimally invasive molecular tests. As such, the review will cover molecular and physiologic biomarkers but leave out histologic and imaging studies. This strategy for biomarker development is summarized in a flowchart adapted from the phased approach provided by the NCI’s early detection research network (Fig. 1) [7].

Fig. 1.

Phases of biomarker development for early detection of pancreatic cancer. Created with BioRender.com.

Fig. 1.

Phases of biomarker development for early detection of pancreatic cancer. Created with BioRender.com.

Close modal

Specimen Collection and Reporting

Since PDAC is largely asymptomatic in earlier stages, any biomarker aiding in early detection would be considered a screening marker [8]. Regardless if the biomarker is tissue-based, blood-based, radiologic, or defined by clinical characteristics, key considerations include sufficient clinical annotation, the inclusion of both non-cancer cases and non-operable stages, and a clear interconnection between the bioinformatics and the specimen itself [9]. To aid researchers in acquiring or utilizing biospecimens, several best practice guidelines exist. The Consortium for the study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer (CPDPC) has developed comprehensive guidelines covering most specimen types and sampling methods (blood, urine, saliva, stool, pancreatic/duodenal juice, and tissue) [10], but standard operating procedures for biobanking of cyst fluid acquisition of PDAC precursor lesions are lacking. Pancreatic cancer tissue is good at maintaining DNA and RNA integrity after prolonged ischemia during handling, while protein and posttranslational modifications are more sensitive [11]. To enhance the reporting of biospecimen-related research and diagnostic accuracy studies, adherence to the Biospecimen Reporting for Improved Study Quality (BRISQ) [12] and the Standards for Reporting of Diagnostic Accuracy Studies (STARD) [13] criteria is recommended.

Blood-Based Markers

The two main configurations for mass spectrometry are liquid chromatography-tandem MS and gas-chromatography MS, with liquid chromatography-tandem MS being the current primary modality for proteomic and metabolomic discovery in PDAC [14]. Residing on mucins of cancer cells, the glycoprotein complex CA 19-9 or sialylated Lewis antigen remains the only blood-based biomarker used in clinical practice, mainly as indication for surgery in pancreatic cystic lesions and for disease monitoring [15]. Baseline levels vary wildly in the general population [16], and elevated levels are seen in many benign conditions, with 10% of patients unable to synthesize it [17]. Preclinically, CA 19-9 functions as an anchor marker in protein panel studies and a reference for overall biomarker performance in PDAC [18, 19]. Mucins are glycoproteins where aberrant and de novo production may closely follow pancreatic tumor progression. Secretory MUC5AC [20] and MUC4 [21] mucins retain high expression through PanIN-3 to invasive disease, but their detection is highly dependent on antigen retrieval methods, and serum concentrations are lower in precancerous disease [22]. Other well-performing glycoproteins are thrombospondins and intercellular adhesion molecules, THBS1 and ICAM-1, oncosuppressive proteins that are downregulated in PDAC [23, 24]. THBS1 and ICAM-1 have good discriminatory power compared to both benign and healthy controls [25]. Furthermore, matrix metalloproteinases are often implicated in PDAC tumor initiation [26], particularly MMP7 which is also expressed in preneoplastic disease [27] and has good diagnostic performance [28]. Recently, MMP2 was reported as a key circulating protease in PDAC and a protease activity-based assay using a magnetic nanosensor was developed, termed PAC-MANN [29]. The assay combined with CA 19-9 was able to detect stage 1 PDAC with 85% sensitivity at 96% specificity. There have also been reports on oncogenic proteases such as cathepsins and urokinase plasminogen activator receptors [30, 31]. In addition, metabolic signatures have demonstrated improved performance compared to CA 19-9 alone in excluding PDAC in a prospective real-world cohort (METAPAC study) [32]. Furthermore, studies of metabolic pathways have generated lipid panels that, combined with CA 19-9, show good ability to differ between PDAC, healthy controls and chronic pancreatitis [33].

Urine Markers

A challenge with urinary biomarkers is high sample variability due to time of day and degradation, information that may not be readily available in the biobank registry [34]. Promisingly, well-performing micro-RNA markers are equivalent between serum and urine [35], and comprehensive predictive tools such as PancRISK have been developed from urinary biomarkers [36].

Liquid Biopsies

Direct tumor constructs, termed “liquid biopsies,” include cell-free plasma DNA/RNA (cfDNA/RNA), circulating tumor cells (CTCs), circulating tumor DNA [37], exosomes [38‒40], and noncoding small RNA, such as circular RNAs [41], microRNAs [42], and transfer RNA-derived RNAs [43]. cfDNA demonstrates low concentrations of point-driver-gene mutations in early PDAC, and clonal hematopoiesis creates a high biological noise [44]. Instead, detection of tumor-specific methylation patterns of cfDNA may be more sensitive, also in PanIN lesions and early PDAC [45]. A recent meta-analysis demonstrated a correlation between peripheral blood CTCs with PDAC stage and survival [46], which could enable detection in earlier invasive disease. Still, the detection rate of CTCs is far lower in precancerous lesions [47].

Sequencing Methods

Next-generation sequencing (NGS) is used for biomarker exploration of tumor constructs in PDAC. Due to low tumor cellularity, employing NGS to PDAC blood samples requires highly sensitive methods. Current NGS methods applied to PDAC include Illumina’s MiSeq, Ion Torrent, Pacific Biosciences, and Oxford Nanopore Technologies (ONT). The long turnaround time and high cost of Illumina sequencing would suggest Ion Torrent methods to be more suited for developing clinical tests. However, few studies targeting PDAC are conducted with Ion Torrent and have mainly been further developments of the platform for pancreatic circulating tumor DNA detection [48]. ONT, as with Ion Torrent, is also hampered by reduced multiplexing capabilities. Still, as ONT provides a shorter gap between the bench and clinic, recent attempts at adequate miRNA detection using ONT show promise for developing more portable liquid biopsy-based tests [35]. Pacific Biosciences has primarily been used in gut microbiome studies of PDAC [49], and MiSeq suffers from high DNA input requirement [50]. Newer third-generation PCR amplification has been given attention, such as multiplexed digital-droplet PCR. It has low turnaround time, highly multiplexed detection, and point-of-care testing capabilities [51], and is frequently applied in miRNA studies for PDAC early detection [52, 53].

Affinity-Based Proteomics

Researchers might use external high-throughput affinity-based biomarker platforms by combining sequencing technologies with proteomic discovery. The Olink platform (Olink Bioscience, Uppsala, Sweden) utilizes proximity extension assays with antibody-conjugated oligonucleotides that hybridize upon target binding and are extended by DNA polymerase for protein quantification [54]. Based on preexisting libraries, thousands of proteins can be detected simultaneously, exceeding the discovery rates of the largest microarray platforms [55]. Published PDAC studies have utilized the “Immuno-oncology” [56], the “Oncology II” [57] and the “Explore 3072” panels [58]. As antibodies are bioreceptors with long half-lives, they create high background noise, lowering the discovery rate. The SomaScan (SomaLogic, Boulder, CR, USA) utilizes short, enriched oligonucleotides (slow-off rate-modified aptamers) treated with protecting groups such as benzene or 2-naphthyl for enhanced plasma stability. SOMAs functionally resemble antibodies but have shorter half-lives and combine with fluorophores for photometric analysis in complex protein samples. Multiplatform studies combining MS, SomaScan, and Olink may expand protein discovery rates even further [59, 60].

Multi-Omics Approaches

While these studies are more focused on etiology of significantly expressed biomarkers, they also give the possibility to conduct purely register-based discovery studies. For example, a proteome-wide association study utilized data from the ARIC and the INTERVAL studies (which in turn are based on the SomaLogic assay) to infer 16 novel proteins not previously reported [61].

Performance Metrics

Upon biomarker discovery, researchers must determine which combinations carry the best performance. The method, often termed “feature selection,” can be done via Monte Carlo optimization [62] or LASSO regression [63]. Consensus regarding PDAC biomarker performance is the need for high specificity, specifically over 98% [64], given the low pretest probability.

Once a candidate biomarker or biomarker signature is identified, a targeted assay platform is developed based on the intended use. Analytical validation involves evaluation of performance metrics of the assay, including sensitivity, specificity, reproducibility, linearity, and dynamic range. Standard operating procedures need to be established. Reference samples from PDAC patients and controls should be analyzed to benchmark results.

The clinical validation step assesses the clinical relevance and utility of the biomarker or biomarker signature.

Retrospective Evaluation

The initial validation phase typically involves case-control analysis of established cases and controls, and retrospective evaluation in a cohort from prospectively collected samples. Controls should include confounding disease states, such as pancreatitis and benign biliary inflammation. Performance results are usually better on the initial patient sample, so cross-validation is used to finalize the prediction model and prevent over-fitting. Traditional ELISA is commonly used for absolute protein quantification after the discovery phase [18, 65]. For liquid biopsy approaches, researchers with tissue-based sequencing have to ensure that the intended sequencing method also is able to detect tumor constructs in noninvasive samples if transitioning to the validation phase [66].

Study Design

For assistance in study design, the protocol from the prospective-specimen-collection retrospective-blinded evaluation (PRoBE) method is preferred. It involves prespecifying the clinical setting, benchmarks, the population at risk, target lesions, and how to ascertain the outcome [8]. Biomarker panel performance is often measured in sensitivity at 98% specificity in conjunction with area under the curve (AUC) metrics to report overall performance, but there have been proposals to use precision-recall curve (AUCPR) to account for class imbalances between PDAC and the control groups. However, recent evidence maintains that regular AUC is the preferred performance metric [67]. Benchmark PDAC performance values from EDRN include an AUC >0.85 and a negative predictive value at or above 99% [68]. Importantly, time-dependent performance metrics are biomarker-dependent in PDAC [69] and could be misleading if used as benchmarks in this phase of development.

Populations at Risk

For pancreatic cancer, five distinct population groups exist for targeting by early detection: (1) patients with pathogenic germline variants, (2) familial pancreatic cancer, (3) symptomatic individuals reporting weight loss combined with new-onset diabetes and other worrisome features (according to NG12 NICE guidelines) [70], (4) patients with chronic pancreatitis, and (5) patients with mucinous cystic neoplasms [71]. Groups 1 and 2 termed high-risk individuals (HRIs) in the literature [16]. In 2019, the US Preventive Services Task Force maintained its recommendation against general population screening, but only in relation to the current performance of the radiologic surveillance strategy for HRIs [16]. Interestingly, sporadic cases with similar radiologic features harbor lower risk lesions upon resection and histological evaluation, while new-onset diabetes bears less relative impact on the likelihood of developing PDAC in familial cases [72]. Hence, from a biological standpoint, HRI surveillance studies do not translate well to sporadic PDAC.

Target Lesions

Imaging is used to confirm the presence of the target lesion. The International Cancer of the Pancreas Screening (CAPS) Consortium defined successful target lesions in HRIs as PanIN3, PanIN with high-grade dysplasia and stage I PDAC [73]. Existing strategies for HRIs yield 5 to 8 target lesions per 1,000 person-years [16]. Concerningly, many prospective biomarker studies fail to include PanIN lesions [16, 74].

Pre-Diagnostic Samples

Researchers should utilize cohorts that recruit PDAC target populations in the clinical setting specified in the PRoBE protocol. This entails pre-diagnostic samples from large cohorts that are largely serum-based, and these could include the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort [75], or ERDN’s own reference set [76], among others.

Tumor Progression

A challenge with pancreatic cancer is the slow progression, modeled to take 7–12 years until metastatic disease [4, 77]. Some PanINs also progress rapidly and undergo multiple simultaneous genomic alterations, mainly by chromosome fragmentation [78]. The sudden appearance of high-stage tumors between the 12-month image study intervals in HRI studies has been attributed to this phenomenon [79]. Ideally, the performance of the biomarkers should not be tied to tumor progression outside of the target lesion range. To evaluate this, researchers should conduct subgroup analyses in the pre-diagnostic cohort based on the time between specimen acquisition and target lesion diagnosis. This is to avoid having to include time-dependent outcome metrics further down the line, for which benchmark values are unclear.

Prospective Evaluation

In transitioning to prospective evaluation in high-risk groups, several important questions must be answered. What should the screening interval be? Is the intervention cost-effective? And most importantly, is there clinical utility? These questions may only fully be answered as the prospective trial is underway, but many prior studies do provide indices [80]. In fact, estimation of the clinically detectable time, e.g., sojourn time, has been invaluable in guiding screening intervals for breast cancer in different age groups [81]. A few studies have calculated average sojourn times for PDAC by hyperglycemic periods and counterfactual analysis, and researchers should complement their own prospective study with their target population’s estimated sojourn times [82]. In the following “penultimate period,” large-scale population studies are set up to evaluate the actual impact of the screening measure. To determine if the “benefit outweighs the cost” in the population, researchers must decide on how many nontarget lesions need to be worked up to detect one target lesion. These threshold values will vary considerably and not only with the disease prevalence of the population at risk, but also with geographic resources, and should not come into play until the last development phases of the biomarker test.

Clinical Trials

Several ongoing clinical trials are investigating biomarkers for the early detection of PDAC. Table 1 provides a summary of selected studies.

Table 1.

Ongoing clinical trials and studies on biomarkers for early detection of PDAC

Trial nameDescriptionCountryBiomarkersReference
A Study of Blood Based Biomarkers for Pancreas Adenocarcinoma Development of biomarkers for the early detection, surveillance, and monitoring of PDAC USA Proteins and proteases, functional DNA repair assays, exosomes, stromal elements, circular RNAs, and ctDNA NCT03334708 
ADEPTS Accelerated Diagnosis of neuroendocrine and Pancreatic TumourS UK Blood, urine, and tissue specimens Pereira et al. [83] (2020) 
ASCEND-PANCREATIC AssesSment of Early-deteCtion basEd oN liquiD Biopsy in PANCREATIC Cancer China cfDNA methylation, ctDNA mutation, serum protein markers, and blood miRNA markers NCT05556603 
DAYBREAK iDentification and vAlidation Model of Liquid biopsY Based cfDNA Methylation and pRotEin biomArKers for Pancreatic Cancer China cfDNA methylation, serum protein markers, and blood miRNA markers NCT05495685 
DETECT Evaluation of a Mixed Meal Test for Diagnosis and Characterization of PancrEaTogEniC DiabeTes Secondary to Pancreatic Cancer and Chronic Pancreatitis USA PP, glucose, c-peptide, insulin, glucagon, GLP-1, and GIP levels Hart et al. [84] (2018) 
ENDPAC Enriching New-onset Diabetes for Pancreatic Cancer USA Age, weight change, and blood glucose (ENDPAC risk score) Chari et al. [85] (2022) 
EUROPAC European Registry of Hereditary Pancreatic Diseases UK Genomic map Boughey et al. [86] (2025) 
ExoLuminate Observational Registry Study to Assess Exo-PDAC Assay Performance for Detection of PDAC in High-Risk or Clinically Suspicious Patients USA EVs isolated from blood plasma (ExoVerita) NCT05625529 
IMAGene Development of a new algorithm to integrate clinical, omics, DNA methylation biomarkers, and epidemiological data for early detection of pancreatic cancer in high-risk individuals France, Italy, Romania, Spain CPRA, including epigenetic biomarkers profiles NCT06334458 
PANLIPSY Early detection of pancreatic cancer by liquid biopsy France CTCs, ctDNA, EVs, circulating immune system, circulating cell-free nucleosomes, proteins, and microbiota Bardol et al. [87] (2024) 
PANXEON PANcreatic cancer Exosome Early detectiON USA, Japan, Republic of Korea 5 cell-free and 8 exosome-miRNAs in plasma samples NCT06388967 
PRECEDE Pancreatic Cancer Early Detection Consortium USA Pathogenic germline variants in pancreatic cancer predisposition genes Zogopoulos et al. [88] (2024) 
PREPAIRD Personalized Surveillance for Early Detection of Pancreatic Cancer in High Risk Individuals Norway ctDNA analysis and protein biomarkers NCT05740111 
U01-Biomarkers for Noninvasive and Early Detection of Pancreatic Cancer Observational, biospecimen collection protocol to develop a bank of pancreatic cancer tissue and normal tissue USA Cell-free and exosomal-miRNA biomarkers using small RNA-Seq in matched tissue and plasma from different cohorts NCT03886571 
UK-EDI United Kingdom Early Detection study UK Blood biomarkers for differentiating between type 3c and type 2 diabetes Oldfield et al. [89] (2022) 
UroPanc Study Early detection of PDAC using a panel of biomarkers UK Urinary biomarker panel (LYVE1, REG1B, TFF1), and affiliated PancRISK score alone or in combination with plasma CA 19-9 NCT04449406 
VAPOR Volatile organic compound Assessment in Pancreatic ductal adenOcaRcinoma UK Volatile organic compounds in exhaled breath NCT05727020 
Trial nameDescriptionCountryBiomarkersReference
A Study of Blood Based Biomarkers for Pancreas Adenocarcinoma Development of biomarkers for the early detection, surveillance, and monitoring of PDAC USA Proteins and proteases, functional DNA repair assays, exosomes, stromal elements, circular RNAs, and ctDNA NCT03334708 
ADEPTS Accelerated Diagnosis of neuroendocrine and Pancreatic TumourS UK Blood, urine, and tissue specimens Pereira et al. [83] (2020) 
ASCEND-PANCREATIC AssesSment of Early-deteCtion basEd oN liquiD Biopsy in PANCREATIC Cancer China cfDNA methylation, ctDNA mutation, serum protein markers, and blood miRNA markers NCT05556603 
DAYBREAK iDentification and vAlidation Model of Liquid biopsY Based cfDNA Methylation and pRotEin biomArKers for Pancreatic Cancer China cfDNA methylation, serum protein markers, and blood miRNA markers NCT05495685 
DETECT Evaluation of a Mixed Meal Test for Diagnosis and Characterization of PancrEaTogEniC DiabeTes Secondary to Pancreatic Cancer and Chronic Pancreatitis USA PP, glucose, c-peptide, insulin, glucagon, GLP-1, and GIP levels Hart et al. [84] (2018) 
ENDPAC Enriching New-onset Diabetes for Pancreatic Cancer USA Age, weight change, and blood glucose (ENDPAC risk score) Chari et al. [85] (2022) 
EUROPAC European Registry of Hereditary Pancreatic Diseases UK Genomic map Boughey et al. [86] (2025) 
ExoLuminate Observational Registry Study to Assess Exo-PDAC Assay Performance for Detection of PDAC in High-Risk or Clinically Suspicious Patients USA EVs isolated from blood plasma (ExoVerita) NCT05625529 
IMAGene Development of a new algorithm to integrate clinical, omics, DNA methylation biomarkers, and epidemiological data for early detection of pancreatic cancer in high-risk individuals France, Italy, Romania, Spain CPRA, including epigenetic biomarkers profiles NCT06334458 
PANLIPSY Early detection of pancreatic cancer by liquid biopsy France CTCs, ctDNA, EVs, circulating immune system, circulating cell-free nucleosomes, proteins, and microbiota Bardol et al. [87] (2024) 
PANXEON PANcreatic cancer Exosome Early detectiON USA, Japan, Republic of Korea 5 cell-free and 8 exosome-miRNAs in plasma samples NCT06388967 
PRECEDE Pancreatic Cancer Early Detection Consortium USA Pathogenic germline variants in pancreatic cancer predisposition genes Zogopoulos et al. [88] (2024) 
PREPAIRD Personalized Surveillance for Early Detection of Pancreatic Cancer in High Risk Individuals Norway ctDNA analysis and protein biomarkers NCT05740111 
U01-Biomarkers for Noninvasive and Early Detection of Pancreatic Cancer Observational, biospecimen collection protocol to develop a bank of pancreatic cancer tissue and normal tissue USA Cell-free and exosomal-miRNA biomarkers using small RNA-Seq in matched tissue and plasma from different cohorts NCT03886571 
UK-EDI United Kingdom Early Detection study UK Blood biomarkers for differentiating between type 3c and type 2 diabetes Oldfield et al. [89] (2022) 
UroPanc Study Early detection of PDAC using a panel of biomarkers UK Urinary biomarker panel (LYVE1, REG1B, TFF1), and affiliated PancRISK score alone or in combination with plasma CA 19-9 NCT04449406 
VAPOR Volatile organic compound Assessment in Pancreatic ductal adenOcaRcinoma UK Volatile organic compounds in exhaled breath NCT05727020 

cfDNA, cell-free DNA; ctDNA, circulating tumor DNA; EV, extracellular vesicles; PDAC, pancreatic ductal adenocarcinoma; CPRA, comprehensive cancer risk prediction algorithm.

The final phase of biomarker development is clinical implementation, which requires regulatory approval from authorities such as US Food and Drug Administration (FDA) or the European Medicines Agency (EMA). Guideline committees such as NCCN and ASCO and key opinion leaders must be engaged for the inclusion of the biomarker in clinical practice recommendations. Post-market surveillance and cost-effectiveness analysis of the biomarker should be performed.

Out of thousands of biomarkers entering the validation phase, very few get approved for clinical use. Biomarkers intended for clinical use must demonstrate not only analytical and clinical validity, but also clinical utility, which means that their use leads to improved clinical decision-making and better results for the patients. Here, we have outlined what we believe to be the most important considerations for molecular biomarkers from bench to clinical practice for early detection of PDAC. To improve clinical translation, researchers should clear several tangible hurdles before moving onto the next phase of testing.

The authors have no conflicts of interest to declare.

This work was supported by the Swedish Cancer Society, the Swedish Research Council, the Crafoord Foundation, the Ingrid and Sverker Persson Foundation, and Regional Research Support as well as ALF funding from Region Skåne.

Axel Bengtsson: conceptualization, investigation, and writing – original draft. Roland Andersson, Johan Linders, and Aiste Gulla: writing – review and editing. Daniel Ansari: conceptualization, writing – review and editing, and supervision.

1.
Santucci
C
,
Mignozzi
S
,
Malvezzi
M
,
Boffetta
P
,
Collatuzzo
G
,
Levi
F
, et al
.
European cancer mortality predictions for the year 2024 with focus on colorectal cancer
.
Ann Oncol
.
2024
;
35
(
3
):
308
16
.
2.
Siegel
RL
,
Giaquinto
AN
,
Jemal
A
.
Cancer statistics, 2024
.
CA Cancer J Clin
.
2024
;
74
(
1
):
12
49
.
3.
Storz
P
.
Acinar cell plasticity and development of pancreatic ductal adenocarcinoma
.
Nat Rev Gastroenterol Hepatol
.
2017
;
14
(
5
):
296
304
.
4.
Connor
AA
,
Gallinger
S
.
Pancreatic cancer evolution and heterogeneity: integrating omics and clinical data
.
Nat Rev Cancer
.
2022
;
22
(
3
):
131
42
.
5.
Matsuda
Y
,
Furukawa
T
,
Yachida
S
,
Nishimura
M
,
Seki
A
,
Nonaka
K
, et al
.
The prevalence and clinicopathological characteristics of high-grade pancreatic intraepithelial neoplasia: autopsy study evaluating the entire pancreatic parenchyma
.
Pancreas
.
2017
;
46
(
5
):
658
64
.
6.
Bengtsson
A
,
Andersson
R
,
Ansari
D
.
The actual 5-year survivors of pancreatic ductal adenocarcinoma based on real-world data
.
Sci Rep
.
2020
;
10
(
1
):
16425
.
7.
Srivastava
S
,
Wagner
PD
.
The early detection research network: a national infrastructure to Support the discovery, development, and validation of cancer biomarkers
.
Cancer Epidemiol Biomarkers Prev
.
2020
;
29
(
12
):
2401
10
.
8.
Pepe
MS
,
Feng
Z
,
Janes
H
,
Bossuyt
PM
,
Potter
JD
.
Pivotal evaluation of the accuracy of a biomarker used for classification or prediction: standards for study design
.
J Natl Cancer Inst
.
2008
;
100
(
20
):
1432
8
.
9.
Balarajah
V
,
Ambily
A
,
Dayem Ullah
AZ
,
Imrali
A
,
Dowe
T
,
Al-Sarireh
B
, et al
.
Pancreatic cancer tissue banks: where are we heading
.
Future Oncol
.
2016
;
12
(
23
):
2661
3
.
10.
Fisher
WE
,
Cruz-Monserrate
Z
,
McElhany
AL
,
Lesinski
GB
,
Hart
PA
,
Ghosh
R
, et al
.
Standard operating procedures for biospecimen collection, processing, and storage: from the consortium for the study of chronic pancreatitis, diabetes, and pancreatic cancer
.
Pancreas
.
2018
;
47
(
10
):
1213
21
.
11.
Peng
J
,
Liu
L
,
Huang
D
,
Chen
H
,
Dai
M
,
Guo
J
, et al
.
Impact of ischemia on sample quality of human pancreatic tissues
.
Pancreatology
.
2020
;
20
(
2
):
265
77
.
12.
Moore
HM
,
Kelly
AB
,
Jewell
SD
,
McShane
LM
,
Clark
DP
,
Greenspan
R
, et al
.
Biospecimen Reporting for Improved Study Quality (BRISQ)
.
J Proteome Res
.
2011
;
10
(
8
):
3429
38
.
13.
Cohen
JF
,
Korevaar
DA
,
Altman
DG
,
Bruns
DE
,
Gatsonis
CA
,
Hooft
L
, et al
.
STARD 2015 guidelines for reporting diagnostic accuracy studies: explanation and elaboration
.
BMJ Open
.
2016
;
6
(
11
):
e012799
.
14.
Ge
P
,
Luo
Y
,
Chen
H
,
Liu
J
,
Guo
H
,
Xu
C
, et al
.
Application of mass spectrometry in pancreatic cancer translational research
.
Front Oncol
.
2021
;
11
:
667427
.
15.
Nakisa
A
,
Sempere
LF
,
Chen
X
,
Qu
LT
,
Woldring
D
,
Crawford
HC
, et al
.
Tumor-Associated carbohydrate antigen 19-9 (CA 19-9), a promising target for antibody-based detection, diagnosis, and immunotherapy of cancer
.
ChemMedChem
.
2024
;
19
(
24
):
e202400491
.
16.
Bogdanski
AM
,
van Hooft
JE
,
Boekestijn
B
,
Bonsing
BA
,
Wasser
M
,
Klatte
DCF
, et al
.
Aspects and outcomes of surveillance for individuals at high-risk of pancreatic cancer
.
Fam Cancer
.
2024
;
23
(
3
):
323
39
.
17.
Lee
T
,
Teng
TZJ
,
Shelat
VG
.
Carbohydrate antigen 19-9 - tumor marker: past, present, and future
.
World J Gastrointest Surg
.
2020
;
12
(
12
):
468
90
.
18.
Capello
M
,
Bantis
LE
,
Scelo
G
,
Zhao
Y
,
Li
P
,
Dhillon
DS
, et al
.
Sequential validation of blood-based protein biomarker candidates for early-stage pancreatic cancer
.
J Natl Cancer Inst
.
2017
;
109
(
4
):
djw266
.
19.
Kim
H
,
Kang
KN
,
Shin
YS
,
Byun
Y
,
Han
Y
,
Kwon
W
, et al
.
Biomarker panel for the diagnosis of pancreatic ductal adenocarcinoma
.
Cancers
.
2020
;
12
(
6
):
1443
.
20.
Kaur
S
,
Smith
LM
,
Patel
A
,
Menning
M
,
Watley
DC
,
Malik
SS
, et al
.
A combination of MUC5AC and CA19-9 improves the diagnosis of pancreatic cancer: a multicenter study
.
Am J Gastroenterol
.
2017
;
112
(
1
):
172
83
.
21.
Sierzega
M
,
Młynarski
D
,
Tomaszewska
R
,
Kulig
J
.
Semiquantitative immunohistochemistry for mucin (MUC1, MUC2, MUC3, MUC4, MUC5AC, and MUC6) profiling of pancreatic ductal cell adenocarcinoma improves diagnostic and prognostic performance
.
Histopathology
.
2016
;
69
(
4
):
582
91
.
22.
Li
N-S
,
Lin
W-L
,
Hsu
Y-P
,
Chen
Y-T
,
Shiue
Y-L
,
Yang
H-W
.
Combined detection of CA19–9 and MUC1 using a colorimetric immunosensor based on magnetic gold nanorods for ultrasensitive risk assessment of pancreatic cancer
.
ACS Appl Bio Mater
.
2019
;
2
(
11
):
4847
55
.
23.
Sawada
T
,
Kimura
K
,
Nishihara
T
,
Onoda
N
,
Teraoka
H
,
Yamashita
Y
, et al
.
TGF-beta1 down-regulates ICAM-1 expression and enhances liver metastasis of pancreatic cancer
.
Adv Med Sci
.
2006
;
51
:
60
5
.
24.
Laklai
H
,
Laval
S
,
Dumartin
L
,
Rochaix
P
,
Hagedorn
M
,
Bikfalvi
A
, et al
.
Thrombospondin-1 is a critical effector of oncosuppressive activity of sst2 somatostatin receptor on pancreatic cancer
.
Proc Natl Acad Sci U S A
.
2009
;
106
(
42
):
17769
74
.
25.
Jenkinson
C
,
Elliott
VL
,
Evans
A
,
Oldfield
L
,
Jenkins
RE
,
O'Brien
DP
, et al
.
Decreased serum thrombospondin-1 levels in pancreatic cancer patients up to 24 Months prior to clinical diagnosis: association with diabetes mellitus
.
Clin Cancer Res
.
2016
;
22
(
7
):
1734
43
.
26.
Knapinska
AM
,
Estrada
CA
,
Fields
GB
.
The roles of matrix metalloproteinases in pancreatic cancer
.
Prog Mol Biol Transl Sci
.
2017
;
148
:
339
54
.
27.
Crawford
HC
,
Scoggins
CR
,
Washington
MK
,
Matrisian
LM
,
Leach
SD
.
Matrix metalloproteinase-7 is expressed by pancreatic cancer precursors and regulates acinar-to-ductal metaplasia in exocrine pancreas
.
J Clin Investig
.
2002
;
109
(
11
):
1437
44
.
28.
Yablecovitch
D
,
Nadler
M
,
Ben-Horin
S
,
Picard
O
,
Yavzori
M
,
Fudim
E
, et al
.
Serum matrix metalloproteinase-7, Syndecan-1, and CA 19-9 as a biomarker panel for diagnosis of pancreatic ductal adenocarcinoma
.
Cancer Med
.
2024
;
13
(
17
):
e70144
.
29.
Montoya Mira
JL
,
Quentel
A
,
Patel
RK
,
Keith
D
,
Sousa
M
,
Minnier
J
, et al
.
Early detection of pancreatic cancer by a high-throughput protease-activated nanosensor assay
.
Sci Transl Med
.
2025
;
17
(
785
):
eadq3110
.
30.
Kalubowilage
M
,
Covarrubias-Zambrano
O
,
Malalasekera
AP
,
Wendel
SO
,
Wang
H
,
Yapa
AS
, et al
.
Early detection of pancreatic cancers in liquid biopsies by ultrasensitive fluorescence nanobiosensors
.
Nanomedicine
.
2018
;
14
(
6
):
1823
32
.
31.
Loosen
SH
,
Tacke
F
,
Püthe
N
,
Binneboesel
M
,
Wiltberger
G
,
Alizai
PH
, et al
.
High baseline soluble urokinase plasminogen activator receptor (suPAR) serum levels indicate adverse outcome after resection of pancreatic adenocarcinoma
.
Carcinogenesis
.
2019
;
40
(
8
):
947
55
.
32.
Mahajan
UM
,
Oehrle
B
,
Goni
E
,
Strobel
O
,
Kaiser
J
,
Grützmann
R
, et al
.
Validation of two plasma multimetabolite signatures for patients at risk of or with suspected pancreatic ductal adenocarcinoma (METAPAC) : a prospective, multicentre, investigator-masked, enrichment-design, phase 4 diagnostic study
.
Lancet Gastroenterol Hepatol
.
2025
;
S2468-1253
(
25
):
00056-1
1
.
33.
Wolrab
D
,
Jirásko
R
,
Cífková
E
,
Höring
M
,
Mei
D
,
Chocholoušková
M
, et al
.
Lipidomic profiling of human serum enables detection of pancreatic cancer
.
Nat Commun
.
2022
;
13
(
1
):
124
.
34.
Debernardi
S
,
Blyuss
O
,
Rycyk
D
,
Srivastava
K
,
Jeon
CY
,
Cai
H
, et al
.
Urine biomarkers enable pancreatic cancer detection up to 2 years before diagnosis
.
Int J Cancer
.
2023
;
152
(
4
):
769
80
.
35.
Kanavarioti
A
,
Rehman
MH
,
Qureshi
S
,
Rafiq
A
,
Sultan
M
.
High sensitivity and specificity platform to validate MicroRNA biomarkers in cancer and human diseases
.
Noncoding RNA
.
2024
;
10
(
4
):
42
.
36.
Blyuss
O
,
Zaikin
A
,
Cherepanova
V
,
Munblit
D
,
Kiseleva
EM
,
Prytomanova
OM
, et al
.
Development of PancRISK, a urine biomarker-based risk score for stratified screening of pancreatic cancer patients
.
Br J Cancer
.
2020
;
122
(
5
):
692
6
.
37.
Wu
H
,
Guo
S
,
Liu
X
,
Li
Y
,
Su
Z
,
He
Q
, et al
.
Noninvasive detection of pancreatic ductal adenocarcinoma using the methylation signature of circulating tumour DNA
.
BMC Med
.
2022
;
20
(
1
):
458
.
38.
Yang
KS
,
Im
H
,
Hong
S
,
Pergolini
I
,
Del Castillo
AF
,
Wang
R
, et al
.
Multiparametric plasma EV profiling facilitates diagnosis of pancreatic malignancy
.
Sci Transl Med
.
2017
;
9
(
391
):
eaal3226
.
39.
Nakamura
K
,
Zhu
Z
,
Roy
S
,
Jun
E
,
Han
H
,
Munoz
RM
, et al
.
An exosome-based transcriptomic signature for noninvasive, early detection of patients with pancreatic ductal adenocarcinoma: a multicenter cohort study
.
Gastroenterology
.
2022
;
163
(
5
):
1252
66.e2
.
40.
Yoshioka
Y
,
Shimomura
M
,
Saito
K
,
Ishii
H
,
Doki
Y
,
Eguchi
H
, et al
.
Circulating cancer-associated extracellular vesicles as early detection and recurrence biomarkers for pancreatic cancer
.
Cancer Sci
.
2022
;
113
(
10
):
3498
509
.
41.
Yang
F
,
Liu
DY
,
Guo
JT
,
Ge
N
,
Zhu
P
,
Liu
X
, et al
.
Circular RNA circ-LDLRAD3 as a biomarker in diagnosis of pancreatic cancer
.
World J Gastroenterol
.
2017
;
23
(
47
):
8345
54
.
42.
Lai
X
,
Wang
M
,
McElyea
SD
,
Sherman
S
,
House
M
,
Korc
M
.
A microRNA signature in circulating exosomes is superior to exosomal glypican-1 levels for diagnosing pancreatic cancer
.
Cancer Lett
.
2017
;
393
:
86
93
.
43.
Xue
M
,
Shi
M
,
Xie
J
,
Zhang
J
,
Jiang
L
,
Deng
X
, et al
.
Serum tRNA-derived small RNAs as potential novel diagnostic biomarkers for pancreatic ductal adenocarcinoma
.
Am J Cancer Res
.
2021
;
11
(
3
):
837
48
.
44.
Ben-Ami
R
,
Wang
QL
,
Zhang
J
,
Supplee
JG
,
Fahrmann
JF
,
Lehmann-Werman
R
, et al
.
Protein biomarkers and alternatively methylated cell-free DNA detect early stage pancreatic cancer
.
Gut
.
2024
;
73
(
4
):
639
48
.
45.
Zhao
G
,
Jiang
R
,
Shi
Y
,
Gao
S
,
Wang
D
,
Li
Z
, et al
.
Circulating cell-free DNA methylation-based multi-omics analysis allows early diagnosis of pancreatic ductal adenocarcinoma
.
Mol Oncol
.
2024
;
18
(
11
):
2801
13
.
46.
Pang
TCY
,
Po
JW
,
Becker
TM
,
Goldstein
D
,
Pirola
RC
,
Wilson
JS
, et al
.
Circulating tumour cells in pancreatic cancer: a systematic review and meta-analysis of clinicopathological implications
.
Pancreatology
.
2021
;
21
(
1
):
103
14
.
47.
Rhim
AD
,
Thege
FI
,
Santana
SM
,
Lannin
TB
,
Saha
TN
,
Tsai
S
, et al
.
Detection of circulating pancreas epithelial cells in patients with pancreatic cystic lesions
.
Gastroenterology
.
2014
;
146
(
3
):
647
51
.
48.
Lapin
M
,
Edland
KH
,
Tjensvoll
K
,
Oltedal
S
,
Austdal
M
,
Garresori
H
, et al
.
Comprehensive ctDNA measurements improve prediction of clinical outcomes and enable dynamic tracking of disease progression in advanced pancreatic cancer
.
Clin Cancer Res
.
2023
;
29
(
7
):
1267
78
.
49.
Pourali
G
,
Kazemi
D
,
Chadeganipour
AS
,
Arastonejad
M
,
Kashani
SN
,
Pourali
R
, et al
.
Microbiome as a biomarker and therapeutic target in pancreatic cancer
.
BMC Microbiol
.
2024
;
24
(
1
):
16
.
50.
Quail
MA
,
Smith
M
,
Coupland
P
,
Otto
TD
,
Harris
SR
,
Connor
TR
, et al
.
A tale of three next generation sequencing platforms: comparison of Ion Torrent, Pacific Biosciences and Illumina MiSeq sequencers
.
BMC Genomics
.
2012
;
13
(
1
):
341
.
51.
Broomfield
J
,
Kalofonou
M
,
Bevan
CL
,
Georgiou
P
.
Recent electrochemical advancements for liquid-biopsy nucleic acid detection for point-of-care prostate cancer diagnostics and prognostics
.
Biosensors
.
2024
;
14
(
9
):
443
.
52.
Tavano
F
,
Gioffreda
D
,
Valvano
MR
,
Palmieri
O
,
Tardio
M
,
Latiano
TP
, et al
.
Droplet digital PCR quantification of miR-1290 as a circulating biomarker for pancreatic cancer
.
Sci Rep
.
2018
;
8
(
1
):
16389
.
53.
Mazza
T
,
Gioffreda
D
,
Fontana
A
,
Biagini
T
,
Carella
M
,
Palumbo
O
, et al
.
Clinical significance of circulating miR-1273g-3p and miR-122-5p in pancreatic cancer
.
Front Oncol
.
2020
;
10
:
44
.
54.
Wik
L
,
Nordberg
N
,
Broberg
J
,
Björkesten
J
,
Assarsson
E
,
Henriksson
S
, et al
.
Proximity extension assay in combination with next-generation sequencing for high-throughput proteome-wide analysis
.
Mol Cell Proteomics
.
2021
;
20
:
100168
.
55.
Huang
YJ
,
Frazier
ML
,
Zhang
N
,
Liu
Q
,
Wei
C
.
Reverse-phase protein array analysis to identify biomarker proteins in human pancreatic cancer
.
Dig Dis Sci
.
2014
;
59
(
5
):
968
75
.
56.
Lindgaard
SC
,
Maag
E
,
Sztupinszki
Z
,
Chen
IM
,
Johansen
AZ
,
Jensen
BV
, et al
.
Circulating protein biomarkers for prognostic use in patients with advanced pancreatic ductal adenocarcinoma undergoing chemotherapy
.
Cancers
.
2022
;
14
(
13
):
3250
.
57.
Yu
J
,
Ploner
A
,
Kordes
M
,
Löhr
M
,
Nilsson
M
,
de Maturana
MEL
, et al
.
Plasma protein biomarkers for early detection of pancreatic ductal adenocarcinoma
.
Int J Cancer
.
2021
;
148
(
8
):
2048
58
.
58.
Athanasiou
A
,
Kureshi
N
,
Wittig
A
,
Sterner
M
,
Huber
R
,
Palma
NA
, et al
.
Biomarker discovery for early detection of Pancreatic Ductal Adenocarcinoma (PDAC) using multiplex proteomics technology
.
J Proteome Res
.
2025
;
24
(
1
):
315
22
.
59.
Pietzner
M
,
Wheeler
E
,
Carrasco-Zanini
J
,
Kerrison
ND
,
Oerton
E
,
Koprulu
M
, et al
.
Synergistic insights into human health from aptamer- and antibody-based proteomic profiling
.
Nat Commun
.
2021
;
12
(
1
):
6822
.
60.
Dammer
EB
,
Ping
L
,
Duong
DM
,
Modeste
ES
,
Seyfried
NT
,
Lah
JJ
, et al
.
Multi-platform proteomic analysis of Alzheimer’s disease cerebrospinal fluid and plasma reveals network biomarkers associated with proteostasis and the matrisome
.
Alzheimers Res Ther
.
2022
;
14
(
1
):
174
.
61.
Zhu
J
,
Wu
K
,
Liu
S
,
Masca
A
,
Zhong
H
,
Yang
T
, et al
.
Proteome-wide association study and functional validation identify novel protein markers for pancreatic ductal adenocarcinoma
.
Gigascience
.
2024
;
13
:
giae012
.
62.
Nolen
BM
,
Brand
RE
,
Prosser
D
,
Velikokhatnaya
L
,
Allen
PJ
,
Zeh
HJ
, et al
.
Prediagnostic serum biomarkers as early detection tools for pancreatic cancer in a large prospective cohort study
.
PLoS One
.
2014
;
9
(
4
):
e94928
.
63.
Borgmästars
E
,
Jacobson
S
,
Simm
M
,
Johansson
M
,
Billing
O
,
Lundin
C
, et al
.
Metabolomics for early pancreatic cancer detection in plasma samples from a Swedish prospective population-based biobank
.
J Gastrointest Oncol
.
2024
;
15
(
2
):
755
67
.
64.
Pepe
MS
,
Janes
H
,
Li
CI
,
Bossuyt
PM
,
Feng
Z
,
Hilden
J
.
Early-phase studies of biomarkers: what target sensitivity and specificity values might confer clinical utility
.
Clin Chem
.
2016
;
62
(
5
):
737
42
.
65.
Kim
J
,
Bamlet
WR
,
Oberg
AL
,
Chaffee
KG
,
Donahue
G
,
Cao
XJ
, et al
.
Detection of early pancreatic ductal adenocarcinoma with thrombospondin-2 and CA19-9 blood markers
.
Sci Transl Med
.
2017
;
9
(
398
):
eaah5583
.
66.
Connal
S
,
Cameron
JM
,
Sala
A
,
Brennan
PM
,
Palmer
DS
,
Palmer
JD
, et al
.
Liquid biopsies: the future of cancer early detection
.
J Transl Med
.
2023
;
21
(
1
):
118
.
67.
Richardson
E
,
Trevizani
R
,
Greenbaum
JA
,
Carter
H
,
Nielsen
M
,
Peters
B
.
The receiver operating characteristic curve accurately assesses imbalanced datasets
.
Patterns
.
2024
;
5
(
6
):
100994
.
68.
Liu
Y
,
Kaur
S
,
Huang
Y
,
Fahrmann
JF
,
Rinaudo
JA
,
Hanash
SM
, et al
.
Biomarkers and strategy to detect preinvasive and early pancreatic cancer: state of the field and the impact of the EDRN
.
Cancer Epidemiol Biomarkers Prev
.
2020
;
29
(
12
):
2513
23
.
69.
Udgata
S
,
Takenaka
N
,
Bamlet
WR
,
Oberg
AL
,
Yee
SS
,
Carpenter
EL
, et al
.
THBS2/CA19-9 detecting pancreatic ductal adenocarcinoma at diagnosis underperforms in prediagnostic detection: implications for biomarker advancement
.
Cancer Prev Res
.
2021
;
14
(
2
):
223
32
.
70.
(NICE) NIfHaCE
.
NG12. Suspected cancer: recognition and referral
.
2015
.
71.
European Study Group on Cystic Tumours of the Pancreas
.
European evidence-based guidelines on pancreatic cystic neoplasms
.
Gut
.
2018
;
67
(
5
):
789
804
.
72.
Bogdanski
AM
,
Onnekink
AM
,
Inderson
A
,
Boekestijn
B
,
Bonsing
BA
,
Vasen
HFA
, et al
.
The added value of blood glucose monitoring in high-risk individuals undergoing pancreatic cancer surveillance
.
Pancreas
.
2024
;
53
(
7
):
e566
72
.
73.
Goggins
M
,
Overbeek
KA
,
Brand
R
,
Syngal
S
,
Del Chiaro
M
,
Bartsch
DK
, et al
.
Management of patients with increased risk for familial pancreatic cancer: updated recommendations from the International Cancer of the Pancreas Screening (CAPS) Consortium
.
Gut
.
2020
;
69
(
1
):
7
17
.
74.
Resovi
A
,
Bani
MR
,
Porcu
L
,
Anastasia
A
,
Minoli
L
,
Allavena
P
, et al
.
Soluble stroma-related biomarkers of pancreatic cancer
.
EMBO Mol Med
.
2018
;
10
(
8
):
e8741
.
75.
Riboli
E
,
Hunt
KJ
,
Slimani
N
,
Ferrari
P
,
Norat
T
,
Fahey
M
, et al
.
European Prospective Investigation into Cancer and Nutrition (EPIC): study populations and data collection
.
Public Health Nutr
.
2002
;
5
(
6b
):
1113
24
.
76.
Feng
Z
,
Kagan
J
,
Pepe
M
,
Thornquist
M
,
Ann Rinaudo
J
,
Dahlgren
J
, et al
.
The early detection research network's specimen reference sets: paving the way for rapid evaluation of potential biomarkers
.
Clin Chem
.
2013
;
59
(
1
):
68
74
.
77.
Peters
MLB
,
Eckel
A
,
Mueller
PP
,
Tramontano
AC
,
Weaver
DT
,
Lietz
A
, et al
.
Progression to pancreatic ductal adenocarcinoma from pancreatic intraepithelial neoplasia: results of a simulation model
.
Pancreatology
.
2018
;
18
(
8
):
928
34
.
78.
Pihlak
R
,
Weaver
JMJ
,
Valle
JW
,
McNamara
MG
.
Advances in molecular profiling and categorisation of pancreatic adenocarcinoma and the implications for therapy
.
Cancers
.
2018
;
10
(
1
):
17
.
79.
Henrikson
NB
,
Aiello Bowles
EJ
,
Blasi
PR
,
Morrison
CC
,
Nguyen
M
,
Pillarisetty
VG
, et al
.
Screening for pancreatic cancer: updated evidence report and systematic review for the US preventive Services Task Force
.
JAMA
.
2019
;
322
(
5
):
445
54
.
80.
Sharma
A
,
Kandlakunta
H
,
Nagpal
SJS
,
Feng
Z
,
Hoos
W
,
Petersen
GM
, et al
.
Model to determine risk of pancreatic cancer in patients with new-onset diabetes
.
Gastroenterology
.
2018
;
155
(
3
):
730
9.e3
.
81.
Cheung
LC
,
Albert
PS
,
Das
S
,
Cook
RJ
.
Multistate models for the natural history of cancer progression
.
Br J Cancer
.
2022
;
127
(
7
):
1279
88
.
82.
Park
J
,
Lim
F
,
Prest
M
,
Ferris
JS
,
Aziz
Z
,
Agyekum
A
, et al
.
Quantifying the potential benefits of early detection for pancreatic cancer through a counterfactual simulation modeling analysis
.
Sci Rep
.
2023
;
13
(
1
):
20028
.
83.
Pereira
S
,
Hippisley-Cox
J
,
Timms
J
,
Hsuan
J
,
Fusai
K
,
Williams
N
, et al
.
ADEPTS (accelerated diagnosis of neuroEndocrine and pancreatic TumourS) and EDRA (early diagnosis research alliance)
.
Pancreatology
.
2020
;
20
(
8
):
e14
.
84.
Hart
PA
,
Andersen
DK
,
Mather
KJ
,
Castonguay
AC
,
Bajaj
M
,
Bellin
MD
, et al
.
Evaluation of a mixed meal test for diagnosis and characterization of PancrEaTogEniC DiabeTes secondary to pancreatic cancer and chronic pancreatitis: rationale and methodology for the DETECT study from the consortium for the study of chronic pancreatitis, diabetes, and pancreatic cancer
.
Pancreas
.
2018
;
47
(
10
):
1239
43
.
85.
Chari
ST
,
Maitra
A
,
Matrisian
LM
,
Shrader
EE
,
Wu
BU
,
Kambadakone
A
, et al
.
Early Detection Initiative: a randomized controlled trial of algorithm-based screening in patients with new onset hyperglycemia and diabetes for early detection of pancreatic ductal adenocarcinoma
.
Contemp Clin Trials
.
2022
;
113
:
106659
.
86.
Boughey
A
,
Hopley
P
,
Sarantitis
I
,
Thomas
P
,
Gubacsi
B
,
Jevons
K
, et al
.
European Registry of Hereditary Pancreatic Diseases (EUROPAC): protocol for primary and secondary screening in individuals with inherited pancreatic disease syndromes for pancreatic ductal adenocarcinoma and complications of other pancreatic diseases
.
BMJ Open
.
2025
;
15
(
4
):
e100027
.
87.
Bardol
T
,
Dujon
AM
,
Taly
V
,
Dunyach-Remy
C
,
Lavigne
JP
,
Costa-Silva
B
, et al
.
Early detection of pancreatic cancer by liquid biopsy “PANLIPSY”: a French nation-wide study project
.
BMC Cancer
.
2024
;
24
(
1
):
709
.
88.
Zogopoulos
G
,
Haimi
I
,
Sanoba
SA
,
Everett
JN
,
Wang
Y
,
Katona
BW
, et al
.
The pancreatic cancer early detection (PRECEDE) study is a global effort to drive early detection: baseline imaging findings in high-risk individuals
.
J Natl Compr Canc Netw
.
2024
;
22
(
3
):
158
66
.
89.
Oldfield
L
,
Stott
M
,
Hanson
R
,
Jackson
RJ
,
Reynolds
W
,
Chandran-Gorner
V
, et al
.
United Kingdom Early Detection Initiative (UK-EDI): protocol for establishing a national multicentre cohort of individuals with new-onset diabetes for early detection of pancreatic cancer
.
BMJ Open
.
2022
;
12
(
10
):
e068010
.