Preprint
Review

This version is not peer-reviewed.

Liquid Biopsy for Early Pancreatic Cancer Detection: Why Has It Not Yet Worked?

Submitted:

03 January 2026

Posted:

05 January 2026

You are already at the latest version

Abstract

Despite extensive technological advances and an ever-growing body of literature, liquid biopsy has yet to achieve reliable early detection of pancreatic ductal adenocarcinoma (PDA). Numerous studies have investigated circulating tumor-derived components, including cell-free DNA (cfDNA), cell-free RNA (cfRNA), extracellular vesicles (EVs), and circulating tumor cells (CTCs), primarily using peripheral blood samples; however, their clinical utility for early-stage disease remains limited. The fundamental obstacles are biological rather than purely technical: early PDA and its precursor lesions, such as pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasms (IPMN), are characterized by minimal tumor burden, low levels of nucleic acid shedding, and substantial background signals from non-neoplastic tissues. Increasing analytical complexity through multilayered liquid biopsy approaches, including analyses from pancreas-associated fluid, has not consistently translated into improved diagnostic performance and, in some cases, has amplified issues related to specificity, reproducibility, and interpretability. Moreover, molecular alterations detected in body fluids may reflect clonal expansion without inevitable malignant progression, raising concerns regarding overdiagnosis and clinical decision-making. Pre-analytical variability, lack of standardization, and limited access to tumor-adjacent fluids further hinder clinical implementation. Liquid biopsy should therefore be regarded as a complementary modality rather than a substitute for histopathological diagnosis, with its precise clinical role in early detection still ill-defined. In this review, we critically examine why liquid biopsy has not yet succeeded in early PDA detection, highlighting the key biological, technical, and clinical barriers that must be addressed to move the field beyond exploratory research toward meaningful clinical application.

Keywords: 
;  ;  ;  ;  ;  

1. Introduction

Recently, extensive research has been conducted on liquid biopsy as minimally invasive testing strategy using various body fluid samples, including blood, urine, and saliva, with the aim of enabling early cancer diagnosis. A wide range of tumor-derived targets in body fluids have been investigated for liquid biopsy, such as cell-free DNA (cfDNA), cell-free RNA (cfRNA), circulating tumor cells (CTCs), extracellular vesicles (EVs), and proteins [1,2,3] (Figure 1). In the field of pancreatobiliary diseases, gastrointestinal biofluids, duodenal fluid (DF) and pancreatic juice (PJ) collected during endoscopic procedure, can be analyzed in addition to blood or urine [4].
Despite substantial research efforts and a growing number of published studies, only a limited number of liquid biopsy approaches have progressed toward clinical implementation, and biomarkers that clearly outperform the conventional marker CA19-9 has not yet been established [5]. The underlying reasons are multifactorial and include challenges in achieving sufficient diagnostic accuracy and reproducibility, limited sensitivity for detecting early-stage disease, and pronounced tumor heterogeneity. To address these issues, diverse strategies are currently being explored across multiple disciplines, including advances in analytical technologies, assay design, and specimen selection [6,7,8].
In this review, we focus on the fundamental challenges in biomarker development for the early diagnosis of pancreatic cancer (PDA). We also summarize the current landscape of recent liquid biopsy research and ongoing efforts to overcome these limitations, with a particular emphasis on cfDNA, cfRNA, and EV-based approaches, including our investigative attempts.

2. Liquid Biopsy Using ctDNA: Current Limitations and Strategies to Address Them

The development of pancreatic cancer precursor lesions, such as pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasms (IPMN), is frequently associated with activating mutations in driver genes including KRAS and GNAS [9]. Subsequent inactivation of tumor suppressor genes, including SMAD4, TP53, and CDKN2A, promote progression of these precancerous lesions to invasive pancreatic ductal adenocarcinoma (PDA) [9]. Accordingly, DNA mutation analysis using cfDNA has been regarded as one of the most central targets in liquid biopsy-based approaches for PDA diagnosis [10].
Beyond somatic mutations, numerous PDA-specific DNA methylation markers have been identified, and their combined application has been proposed as a promising strategy for early diagnosis [11,12,13,14]. In addition, recent studies have demonstrated that cfDNA fragmentation patterns, including fragment sizes and end motifs, differs between patients with PDA and healthy individuals. Machine learning models integrating these fragmentomic features have been suggested to contribute to early diagnosis and, potentially, inference of the cell of origin of tumor [15]. Nevertheless, despite the clinical implemented of plasma cfDNA analysis for cancer genome profiling (CGP) tests in Japan, its application remains limited to therapeutic decision-making rather than primary cancer diagnosis [16].
Plasma cfDNA-based liquid biopsy offers several advantages, including minimally invasiveness, ease of sample collection enabling longitudinal monitoring, short turnaround time (TAT), and the ability to capture systemic tumor heterogeneity rather than information from a single lesion. However, cfDNA concentrations in peripheral blood are inherently low, highly fragmented, and susceptible to chemical degradation. Moreover, the proportion of tumor-derived ctDNA within total cfDNA is strongly influenced by tumor size, stage, and the proximity to circulating body fluids, resulting in limited mutation detection rate in early-stages of PDA [17].
Pre-analytical factors further complicate cfDNA analysis. Delays between sample collection and processing, as well as variations in storage conditions, can substantially affect the analytical results. For example, leukocyte lysis during blood storage releases genomic DNA, increasing background DNA levels and reducing variant allele frequency (VAF), thereby impairing the detection of low-frequency mutations [18]. Standardized protocols for plasma separation, storage, processing are therefore essential. Additionally, mutations arising from clonal hematopoiesis of indeterminate potential (CHIP), which increases with aging, represent a major source of false-positives findings. Because hematopoietic cells constitute the primary source of cfDNA is hematopoietic cells, CHIP-associated mutations , including those in DNMT3A and TET2, as well as PDA-relevant genes such as TP53 and BRCA1/2 may be misidentified as tumor-derived alterations [19,20], thereby reducing diagnostic specificity.
Genetic analysis of cfDNA can be broadly categorized into PCR amplification-based detection methods and sequencing-based genomic profiling approaches that enable broader genomic coverage. PCR-based techniques include real-time PCR, digital PCR (dPCR), and the extremely sensitive BEAMing method, which is particularly suited for detecting predefined hotspot mutations. These approaches offer high analytical sensitivity (approximately 0.01% for dPCR and BEAMing), relatively low cost, and simplified workflows; however, they are inherently limited in the number of target regions that can be interrogated simultaneously.
In contrast, sequencing-based cfDNA analysis incorporating molecular barcoding and error-suppression strategies allow for the detection of low-frequency variants at allele frequencies of approximately 0.1–1%, which is substantially lower than those detectable by standard target sequencing approaches without error correction. Compared with PCR-based assays, sequencing-based genomic profiling enables the simultaneous identification of a broader range of genomic alterations, including point mutations and copy number variations across multiple genes, thereby providing a more comprehensive overview of tumor-associated genomic changes [1,21].
To overcome these limitations, various strategies are currently being explored. These include the use of pre-amplification combined with dPCR to enhance sensitivity for low-abundance mutations, as well as multiplexed assay design developed by the authors to maximize information yield from limited sample volumes [22]. In parallel, specialized blood collection tubes capable of stabilizing nucleic acids (e.g., Streck, Becton Dickinson, and Roche) have been introduced, with expanding applications for both cfDNA and cfRNA preservation.

3. Liquid Biopsy Using EV and RNA: Complementary Approaches and Remaining Challenges

As discussed above, cfDNA-based detection of KRAS mutations has intrinsic limitations, particularly in early-stage disease. To complement its diagnostic performance, EVs and non-coding RNAs (ncRNAs) have emerged as alternative and potentially informative targets in liquid biopsy research.
ncRNAs are broadly classified based on a transcript length, small ncRNAs (<200 nucleotides) including microRNAs (miRNAs), and with long non-coding RNAs (lncRNAs) exceeding of 200 nucleotides [23,24]. Among these, miRNAs, typically 21–25 nucleotides in length, have been most extensively investigated as circulating biomarkers [25,26], whereas studies on lncRNAs remain relatively limited [24,27,28]. Several reports have demonstrated the diagnostic potential of miRNAs detected as cfRNA in body fluids. For example, Nakamura et al. validated a 13-miRNA signature for PDA detection, archiving an area under the curve (AUC) of 0.93 [29]. Another study employing machine learning of urinary miRNA combinations reported high diagnostic accuracy for PDA, including early-stage disease [30].
EVs are membrane-bound vesicles secreted by virtually all cell types and are present in diverse body fluids, including blood, urine, breast milk, and saliva. EVs contain a variety of bioactive molecules, such as proteins and nucleic acids, reflecting the molecular characteristics of their cells of origin [31,32]. Recent studies have demonstrated the diagnostic potential of EV-associated markers in pancreatic diseases. For instance, elevated expression of MUC5AC in plasma EVs was shown to distinguish high-grade IPMNs from low-grade IPMNs with high sensitivity and specificity [33].
However, EV-based biomarker development faces substantial technical challenges, particularly regarding isolation methods, purity, and reproducibility. Commonly used EV isolation techniques, such as precipitation and ultracentrifugation, are prone to co-isolation of non-EV contaminants and inter-method variability [31]. To address these issues, we have developed a proprietary EV isolation platform, EViSTEP®, based on immunoprecipitation using antibody-coupled magnetic particles targeting the tetraspanin, combined with a pretreatment reagent that enhances EV–antibody interactions while reducing contaminating proteins [34]. Using this platform, we identified a previously unrecognized lncRNA, HEVEPA, which was significantly upregulated in serum EVs from PDA patients compared to healthy controls and IPMN patients, achieving an AUC of 0.86. Notably, HEVEPA elevation was observed specifically within EVs rather than as cfRNA, underscoring the value of EV-targeted biomarker discovery [34].
Despite these advances, standardization remains a major challenges in cfRNA and EV analyses, particularly with respect to endogenous reference controls. While several stable miRNAs (e.g., miR-149-3p, miR-2861, and miR-4463) have been proposed as cfRNA [35], no consensus has been established for EV-based normalization strategies. In our preliminary analysis, ACTB and B2M demonstrated relatively stable expression within EVs, and further validation studies are ongoing.

4. Recent Application and Studies as Liquid Biopsy

In recent years, several biomarkers have been introduced into clinical practice under health insurance coverage in Japan. Among these, the APOA2 isoform index (APO2-iTQ) has been implemented as an adjunctive marker for PDA diagnosis. Multiple studies have demonstrated significantly reduced APOA2-ATQ/AT levels in patients with PDA, with reported AUC values comparable to or exceeding those of CA19-9 in distinguishing across disease stages [36,37,38,39].
Another promising marker is EphA2-NF, a circulating N-terminal fragment of the receptor tyrosine kinase EphA2, which is frequently overexpressed in PDA cells. EphA2-NF is independent of CA19-9 and have been associated with poor prognosis among patients receiving gemcitabine plus nab-paclitaxel (GnP) treatment, suggesting it potential clinical utility as both a diagnostic and prognostic marker [40].
Recent cfDNA-based studies have reported remarkably high diagnostic performance using artificial intelligence (AI)-driven models integrating cfDNA fragmentomics, copy-number variation (CNV), end motifs, and methylation features, with AUC value approaching 0.992 when compared with healthy controls [15]. In addition, methylation-based approaches such as methylated CpG tandem amplification and sequencing (MCTA-Seq) have identified multiple candidate biomarkers with high diagnostic accuracy [41,42]
Tumor-adjacent fluids have also attracted attention as alternative liquid biopsy sources. Yachida et al. reported high diagnostic accuracy for PDA using KRAS mutation analysis of DF collected after secretin [43], while analysis of S100P protein expression in DF has shown potential utility as a minimally invasive screening approach [44,45].
EV-based biomarker studies have further expanded the liquid biopsy landscape. EV surface protein Glypican-1 (GPC1)-positive EVs, in combination with CD82 and CA19-9, have demonstrated high diagnostic accuracy for differentiating PDA from chronic pancreatitis [46]. As summarized in Table 1, Additional studies have identified EV-associated miRNAs, circRNAs, and lncRNAs with diagnostic potential, some achieving performance comparable to or exceeding that of CA19-9 alone [47,48,49,50,51,52].
Collectively, these studies underscore the extensive global efforts directed toward biomarker discovery and validation. Rather than relying on a single molecular target, integrating multiple biomarkers across different liquid biopsy platforms may offer a more realistic path toward improved diagnostic accuracy. Accordingly, we propose the development and clinical implementation of a cost-effective "Pancreatic Cancer Diagnostic Panel" that combines complementary biomarkers, as conceptually illustrated in Figure 1.

5. Conclusion and Future Perspectives

Despite substantial advances in liquid biopsy technologies, early detection of PDA remains an unresolved clinical challenge, primarily due to biological constraints such as minimal tumor-derived nucleic acid abundance, marked heterogeneity, and the difficulty in discriminating against indolent precursor lesions from those with malignant potential. Increasing analytical sophistication alone has not been sufficient to overcome these limitations, underscoring the need to reconsider the clinical positioning of liquid biopsy. Rather than serving as a stand-alone screening tool, liquid biopsy is more realistically positioned as a complementary modality that supports diagnosis and risk stratification in well-defined clinical settings. Future progress will likely depend on rationally designed diagnostic panels that integrate multiple molecular features across different biofluids with clinical and imaging information, evaluated not only for analytical performance but also for interpretability, feasibility, and cost-effectiveness. Aligning technological innovation with biological insight and clinical need will be essential for the meaningful clinical implementation of liquid biopsy in pancreatic cancer.

Author Contributions

KenjiT, YO and KenzuiT were involved in conceptualization, literature search, interpretation of the data, and drafting of the article. KP, TY, MF and YM performed critical revision of the article and supervision of the study.

Funding

This work funded in part by the Japan Society for the Promotion of Science (JSPS) KAKENHI [Grant Numbers: 24K11058 (to KenjiT)]. KenjiT also received support from the Suzuken Memorial Foundation .

Acknowledgments

The authors thank their colleagues in the Department of Internal Medicine, Asahikawa Medical University; Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health; and Nutrition and Institute of Biomedical Research, Sapporo Higashi Tokushukai Hospital for supporting the review, and H.U. Group Research Institute G.K. for creating a figure.

Conflicts of Interest

KenjiT has received grant support from H.U. Group Research Institute G.K. KenjiT, YO and YM have received grant support from Hitachi High-Tech.

Abbreviations

The following abbreviations are used in this manuscript:
PDA pancreatic ductal adenocarcinoma
cfDNA cell-free DNA
cfRNA cell-free RNA
EVs extracellular vesicles
CTCs circulating tumor cells
PanIN pancreatic intraepithelial neoplasia
IPMN intraductal papillary mucinous neoplasms
DF duodenal fluid
PJ pancreatic juice
CGP cancer genome profiling
TAT turnaround time
VAF frequency
CHIP clonal hematopoiesis of indeterminate potential
dPCR digital PCR
ncRNAs non-coding RNAs
miRNAs microRNAs
lncRNAs long non-coding RNAs
AUC area under the curve
APO2-iTQ APOA2 isoform index
GnP gemcitabine plus nab-paclitaxel
AI artificial intelligence
CNV copy-number variation
MCTA-Seq methylated CpG tandem amplification and sequencing
GPC1 Glypican-1

References

  1. Ignatiadis, M.; Sledge, G.W.; Jeffrey, S.S. Liquid biopsy enters the clinic - implementation issues and future challenges. Nat Rev Clin Oncol 2021, 18, 297–312. [Google Scholar] [CrossRef]
  2. Siravegna, G.; Marsoni, S.; Siena, S.; Bardelli, A. Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol 2017, 14, 531–548. [Google Scholar] [CrossRef]
  3. 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]
  4. Takahashi, K.; Takeda, Y.; Ono, Y.; Isomoto, H.; Mizukami, Y. Current status of molecular diagnostic approaches using liquid biopsy. J Gastroenterol 2023, 58, 834–847. [Google Scholar] [CrossRef]
  5. Fahrmann, J.F.; Schmidt, C.M.; Mao, X.; Irajizad, E.; Loftus, M.; Zhang, J.; Patel, N.; Vykoukal, J.; Dennison, J.B.; Long, J.P.; et al. Lead-Time Trajectory of CA19-9 as an Anchor Marker for Pancreatic Cancer Early Detection. Gastroenterology 2021, 160, 1373–1383 e1376. [Google Scholar] [CrossRef]
  6. Hidalgo, M. Pancreatic cancer. N Engl J Med 2010, 362, 1605–1617. [Google Scholar] [CrossRef]
  7. Kamisawa, T.; Wood, L.D.; Itoi, T.; Takaori, K. Pancreatic cancer. Lancet 2016, 388, 73–85. [Google Scholar] [CrossRef] [PubMed]
  8. Cao, L.; Huang, C.; Cui Zhou, D.; Hu, Y.; Lih, T.M.; Savage, S.R.; Krug, K.; Clark, D.J.; Schnaubelt, M.; Chen, L.; et al. Proteogenomic characterization of pancreatic ductal adenocarcinoma. Cell 2021, 184, 5031–5052 e5026. [Google Scholar] [CrossRef] [PubMed]
  9. Patra, K.C.; Bardeesy, N.; Mizukami, Y. Diversity of Precursor Lesions For Pancreatic Cancer: The Genetics and Biology of Intraductal Papillary Mucinous Neoplasm. Clin Transl Gastroenterol 2017, 8, e86. [Google Scholar] [CrossRef]
  10. 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] [PubMed]
  11. Garcia-Ortiz, M.V.; Cano-Ramirez, P.; Toledano-Fonseca, M.; Aranda, E.; Rodriguez-Ariza, A. Diagnosing and monitoring pancreatic cancer through cell-free DNA methylation: progress and prospects. Biomark Res 2023, 11, 88. [Google Scholar] [CrossRef]
  12. Vrba, L.; Futscher, B.W.; Oshiro, M.; Watts, G.S.; Menashi, E.; Hu, C.; Hammad, H.; Pennington, D.R.; Golconda, U.; Gavini, H.; et al. Liquid biopsy, using a novel DNA methylation signature, distinguishes pancreatic adenocarcinoma from benign pancreatic disease. Clin Epigenetics 2022, 14, 28. [Google Scholar] [CrossRef]
  13. Ono, M.; Ono, Y.; Nakamura, T.; Tsuchikawa, T.; Kuraya, T.; Kuwabara, S.; Nakanishi, Y.; Asano, T.; Matsui, A.; Tanaka, K.; et al. Predictors of Long-Term Survival in Pancreatic Ductal Adenocarcinoma after Pancreatectomy: TP53 and SMAD4 Mutation Scoring in Combination with CA19-9. Ann Surg Oncol 2022, 29, 5007–5019. [Google Scholar] [CrossRef]
  14. Omori, Y.; Ono, Y.; Morikawa, T.; Motoi, F.; Higuchi, R.; Yamamoto, M.; Hayakawa, Y.; Karasaki, H.; Mizukami, Y.; Unno, M.; et al. Serine/Threonine Kinase 11 Plays a Canonical Role in Malignant Progression of KRAS-mutant and GNAS-wild-type Intraductal Papillary Mucinous Neoplasms of the Pancreas. Ann Surg 2021. [Google Scholar] [CrossRef]
  15. Yin, L.; Cao, C.; Lin, J.; Wang, Z.; Peng, Y.; Zhang, K.; Xu, C.; Yang, R.; Zhu, D.; Wang, F.; et al. Development and Validation of a Cell-Free DNA Fragmentomics-Based Model for Early Detection of Pancreatic Cancer. J Clin Oncol 2025, 43, 2863–2874. [Google Scholar] [CrossRef]
  16. Nakamura, Y.; Taniguchi, H.; Ikeda, M.; Bando, H.; Kato, K.; Morizane, C.; Esaki, T.; Komatsu, Y.; Kawamoto, Y.; Takahashi, N.; et al. Clinical utility of circulating tumor DNA sequencing in advanced gastrointestinal cancer: SCRUM-Japan GI-SCREEN and GOZILA studies. Nat Med 2020, 26, 1859–1864. [Google Scholar] [CrossRef]
  17. Song, P.; Wu, L.R.; Yan, Y.H.; Zhang, J.X.; Chu, T.; Kwong, L.N.; Patel, A.A.; Zhang, D.Y. Limitations and opportunities of technologies for the analysis of cell-free DNA in cancer diagnostics. Nat Biomed Eng 2022, 6, 232–245. [Google Scholar] [CrossRef]
  18. Norton, S.E.; Lechner, J.M.; Williams, T.; Fernando, M.R. A stabilizing reagent prevents cell-free DNA contamination by cellular DNA in plasma during blood sample storage and shipping as determined by digital PCR. Clin Biochem 2013, 46, 1561–1565. [Google Scholar] [CrossRef]
  19. Hu, Y.; Ulrich, B.C.; Supplee, J.; Kuang, Y.; Lizotte, P.H.; Feeney, N.B.; Guibert, N.M.; Awad, M.M.; Wong, K.K.; Janne, P.A.; et al. False-Positive Plasma Genotyping Due to Clonal Hematopoiesis. Clin Cancer Res 2018, 24, 4437–4443. [Google Scholar] [CrossRef]
  20. Razavi, P.; Li, B.T.; Brown, D.N.; Jung, B.; Hubbell, E.; Shen, R.; Abida, W.; Juluru, K.; De Bruijn, I.; Hou, C.; et al. High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. Nat Med 2019, 25, 1928–1937. [Google Scholar] [CrossRef]
  21. Vidal, J.; Muinelo, L.; Dalmases, A.; Jones, F.; Edelstein, D.; Iglesias, M.; Orrillo, M.; Abalo, A.; Rodriguez, C.; Brozos, E.; et al. Plasma ctDNA RAS mutation analysis for the diagnosis and treatment monitoring of metastatic colorectal cancer patients. Ann Oncol 2017, 28, 1325–1332. [Google Scholar] [CrossRef]
  22. Maeda, C; Takahashi, O.Y.K. Six-color multiplex digital PCR assays for screening and identification of multiple driver mutations associated with pancreatic carcinogenesis. Clin Chem. 2025, in press. [Google Scholar]
  23. Takahashi, K.; Yan, I.; Haga, H.; Patel, T. Long noncoding RNA in liver diseases. Hepatology 2014, 60, 744–753. [Google Scholar] [CrossRef]
  24. Slack, F.J.; Chinnaiyan, A.M. The Role of Non-coding RNAs in Oncology. Cell 2019, 179, 1033–1055. [Google Scholar] [CrossRef]
  25. Srinivasan, S.; Yeri, A.; Cheah, P.S.; Chung, A.; Danielson, K.; De Hoff, P.; Filant, J.; Laurent, C.D.; Laurent, L.D.; Magee, R.; et al. Small RNA Sequencing across Diverse Biofluids Identifies Optimal Methods for exRNA Isolation. Cell 2019, 177, 446–462 e416. [Google Scholar] [CrossRef]
  26. Nesteruk, K.; Levink, I.J.M.; de Vries, E.; Visser, I.J.; Peppelenbosch, M.P.; Cahen, D.L.; Fuhler, G.M.; Bruno, M.J. Extracellular vesicle-derived microRNAs in pancreatic juice as biomarkers for detection of pancreatic ductal adenocarcinoma. Pancreatology 2022, 22, 626–635. [Google Scholar] [CrossRef]
  27. Takahashi, K.; Yan, I.K.; Kogure, T.; Haga, H.; Patel, T. Extracellular vesicle-mediated transfer of long non-coding RNA ROR modulates chemosensitivity in human hepatocellular cancer. FEBS Open Bio 2014, 4, 458–467. [Google Scholar] [CrossRef]
  28. Yu, S.; Li, Y.; Liao, Z.; Wang, Z.; Wang, Z.; Li, Y.; Qian, L.; Zhao, J.; Zong, H.; Kang, B.; et al. Plasma extracellular vesicle long RNA profiling identifies a diagnostic signature for the detection of pancreatic ductal adenocarcinoma. Gut 2020, 69, 540–550. [Google Scholar] [CrossRef]
  29. Nakamura, K.; Zhu, Z.; Roy, S.; Jun, E.; Han, H.; Munoz, R.M.; Nishiwada, S.; Sharma, G.; Cridebring, D.; Zenhausern, F.; et al. An Exosome-based Transcriptomic Signature for Noninvasive, Early Detection of Patients With Pancreatic Ductal Adenocarcinoma: A Multicenter Cohort Study. Gastroenterology 2022, 163, 1252–1266 e1252. [Google Scholar] [CrossRef]
  30. Baba, S.; Kawasaki, T.; Hirano, S.; Nakamura, T.; Asano, T.; Okazaki, R.; Yoshida, K.; Kawase, T.; Kurahara, H.; Oi, H.; et al. A noninvasive urinary microRNA-based assay for the detection of pancreatic cancer from early to late stages: a case control study. EClinicalMedicine 2024, 78, 102936. [Google Scholar] [CrossRef]
  31. Liang, K.; Liu, F.; Fan, J.; Sun, D.; Liu, C.; Lyon, C.J.; Bernard, D.W.; Li, Y.; Yokoi, K.; Katz, M.H.; et al. Nanoplasmonic Quantification of Tumor-derived Extracellular Vesicles in Plasma Microsamples for Diagnosis and Treatment Monitoring. Nat Biomed Eng 2017, 1. [Google Scholar] [CrossRef]
  32. Kalluri, R.; McAndrews, K.M. The role of extracellular vesicles in cancer. Cell 2023, 186, 1610–1626. [Google Scholar] [CrossRef]
  33. Yang, K.S.; Ciprani, D.; O'Shea, A.; Liss, A.S.; Yang, R.; Fletcher-Mercaldo, S.; Mino-Kenudson, M.; Fernandez-Del Castillo, C.; Weissleder, R. Extracellular Vesicle Analysis Allows for Identification of Invasive IPMN. Gastroenterology 2021, 160, 1345–1358 e1311. [Google Scholar] [CrossRef]
  34. Takahashi, K.; Inuzuka, T.; Shimizu, Y.; Sawamoto, K.; Taniue, K.; Ono, Y.; Asai, F.; Koyama, K.; Sato, H.; Kawabata, H.; et al. Liquid Biopsy for Pancreatic Cancer by Serum Extracellular Vesicle-Encapsulated Long Noncoding RNA HEVEPA. Pancreas 2024, 53, e395–e404. [Google Scholar] [CrossRef]
  35. Yokoi, A.; Matsuzaki, J.; Yamamoto, Y.; Yoneoka, Y.; Takahashi, K.; Shimizu, H.; Uehara, T.; Ishikawa, M.; Ikeda, S.I.; Sonoda, T.; et al. Integrated extracellular microRNA profiling for ovarian cancer screening. Nat Commun 2018, 9, 4319. [Google Scholar] [CrossRef]
  36. Kashiro, A.; Kobayashi, M.; Oh, T.; Miyamoto, M.; Atsumi, J.; Nagashima, K.; Takeuchi, K.; Nara, S.; Hijioka, S.; Morizane, C.; et al. Clinical development of a blood biomarker using apolipoprotein-A2 isoforms for early detection of pancreatic cancer. J Gastroenterol 2024, 59, 263–278. [Google Scholar] [CrossRef]
  37. Honda, K.; Hayashida, Y.; Umaki, T.; Okusaka, T.; Kosuge, T.; Kikuchi, S.; Endo, M.; Tsuchida, A.; Aoki, T.; Itoi, T.; et al. Possible detection of pancreatic cancer by plasma protein profiling. Cancer Res 2005, 65, 10613–10622. [Google Scholar] [CrossRef]
  38. Honda, K.; Kobayashi, M.; Okusaka, T.; Rinaudo, J.A.; Huang, Y.; Marsh, T.; Sanada, M.; Sasajima, Y.; Nakamori, S.; Shimahara, M.; et al. Plasma biomarker for detection of early stage pancreatic cancer and risk factors for pancreatic malignancy using antibodies for apolipoprotein-AII isoforms. Sci Rep 2015, 5, 15921. [Google Scholar] [CrossRef]
  39. Hanada, K.; Shimizu, A.; Tsushima, K.; Kobayashi, M. Potential of Carbohydrate Antigen 19-9 and Serum Apolipoprotein A2-Isoforms in the Diagnosis of Stage 0 and IA Pancreatic Cancer. Diagnostics (Basel) 2024, 14. [Google Scholar] [CrossRef]
  40. Sato, S.; Nakagawa, M.; Terashima, T.; Morinaga, S.; Miyagi, Y.; Yoshida, E.; Yoshimura, T.; Seiki, M.; Kaneko, S.; Ueno, M.; et al. EphA2 Proteolytic Fragment as a Sensitive Diagnostic Biomarker for Very Early-stage Pancreatic Ductal Carcinoma. Cancer Res Commun 2023, 3, 1862–1874. [Google Scholar] [CrossRef]
  41. Hu, W.; Zhao, X.; Luo, N.; Xiao, M.; Feng, F.; An, Y.; Chen, J.; Rong, L.; Yang, Y.; Peng, J. Circulating cell-free DNA methylation analysis of pancreatic cancer patients for early noninvasive diagnosis. Front Oncol 2025, 15, 1552426. [Google Scholar] [CrossRef]
  42. Suehiro, Y.; Suenaga, S.; Kunimune, Y.; Yada, S.; Hamamoto, K.; Tsuyama, T.; Amano, S.; Matsui, H.; Higaki, S.; Fujii, I.; et al. CA19-9 in Combination with Methylated HOXA1 and SST Is Useful to Diagnose Stage I Pancreatic Cancer. Oncology 2022, 100, 674–684. [Google Scholar] [CrossRef]
  43. Yachida, S.; Yoshinaga, S.; Shiba, S.; Urabe, M.; Tanaka, H.; Takeda, Y.; Shimizu, A.; Sakamoto, Y.; Hijioka, S.; Haba, S.; et al. KRAS Mutations in Duodenal Lavage Fluid after Secretin Stimulation for Detection of Pancreatic Cancer. Ann Surg 2025. [Google Scholar] [CrossRef]
  44. Matsunaga, T.; Ohtsuka, T.; Asano, K.; Kimura, H.; Ohuchida, K.; Kitada, H.; Ideno, N.; Mori, Y.; Tokunaga, S.; Oda, Y.; et al. S100P in Duodenal Fluid Is a Useful Diagnostic Marker for Pancreatic Ductal Adenocarcinoma. Pancreas 2017, 46, 1288–1295. [Google Scholar] [CrossRef]
  45. Ideno, N.; Mori, Y.; Nakamura, M.; Ohtsuka, T. Early Detection of Pancreatic Cancer: Role of Biomarkers in Pancreatic Fluid Samples. Diagnostics (Basel) 2020, 10. [Google Scholar] [CrossRef]
  46. 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 2020, 18, 300–310. [Google Scholar] [CrossRef]
  47. Hong, L.; Xu, L.; Jin, L.; Xu, K.; Tang, W.; Zhu, Y.; Qiu, X.; Wang, J. Exosomal circular RNA hsa_circ_0006220, and hsa_circ_0001666 as biomarkers in the diagnosis of pancreatic cancer. J Clin Lab Anal 2022, 36, e24447. [Google Scholar] [CrossRef]
  48. Makler, A.; Narayanan, R.; Asghar, W. An Exosomal miRNA Biomarker for the Detection of Pancreatic Ductal Adenocarcinoma. Biosensors (Basel) 2022, 12. [Google Scholar] [CrossRef]
  49. Makler, A.; Asghar, W. Exosomal miRNA Biomarker Panel for Pancreatic Ductal Adenocarcinoma Detection in Patient Plasma: A Pilot Study. Int J Mol Sci 2023, 24. [Google Scholar] [CrossRef]
  50. Xu, J.; Shi, W.; Zhu, Y.; Zhang, C.; Nagelschmitz, J.; Doelling, M.; Al-Madhi, S.; Mukund Mahajan, U.; Pech, M.; Rose, G.; et al. Human multiethnic radiogenomics reveals low-abundancy microRNA signature in plasma-derived extracellular vesicles for early diagnosis and molecular subtyping of pancreatic cancer. Elife 2025, 14. [Google Scholar] [CrossRef]
  51. Patel, R.D.; Patel, B.; Crnogorac-Jurcevic, T. Extracellular Vesicle-Derived miRNAs as Diagnostic Biomarkers for Pancreatic Ductal Adenocarcinoma: A Systematic Review of Methodological Rigour and Clinical Applicability. Biomark Insights 2025, 20, 11772719251381960. [Google Scholar] [CrossRef] [PubMed]
  52. 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]
Figure 1. Overview of Liquid Biopsy for generating multilayer gene panel.
Figure 1. Overview of Liquid Biopsy for generating multilayer gene panel.
Preprints 192722 g001
Table 1. Recent findings in liquid biopsy research for PDA.
Table 1. Recent findings in liquid biopsy research for PDA.
Analysis target Molecular target Type of clinical sample Potential roles for liquid biopsy for PDA diagnosis Reference
cfDNA cfDNA fragmentation pattern plasma Machine learning models integrating cfDNA fragmentation patterns, including fragment sizes and end motifs. Yin et al (2025)
[15]
cfRNA / EV 13 miRNAs (5 cfRNA and 8 EV RNA) plasma / serum The combination of 13-miRNA signature archived an area under the curve (AUC) of 0.98 in training cohort and 0.93 in validation cohort for PDA detection. Nakamura et al (2022)
[29]
EV EV miRNA-based detection set urine Machine learning of urinary extracellular vesicle miRNA combinations showed high diagnostic accuracy for PDA, including early-stage disease. Baba et al (2024)
[30]
EV MUC5AC plasma MUC5AC in plasma EVs was shown to distinguish high-grade IPMNs from low-grade IPMNs with high sensitivity and specificity. Yang et al (2021)
[33]
EV lncRNA HEVEPA serum HEVEPA expression was upregulated in serum EVs from PDA patients compared to healthy controls and IPMN patients, achieving an AUC of 0.86. Takahashi et al (2024)
[33]
protein APO2-iTQ blood APOA2 isoform index (APO2-iTQ) has been implemented as an adjunctive marker for PDA diagnosis in Japan. Hanada et al. (2024)
[39]
protein EphA2-NF serum EphA2-NF is associated with poor prognosis among patients receiving GnP treatment, and has potential clinical utility as both a diagnostic and prognostic marker. Sato et al. (2023)
[40]
cfDNA methylated CpG tandem amplification plasma Methylation scoring and typing system achieved a sensitivity of 97% and 86% for patients in the discovery and validation cohorts, respectively, with a specificity of 100% in both cohorts for PDA. Hu et al. (2025)
[41]
cfDNA methylated Homeobox A1 (mHOXA1) and methylated somatostatin (mSST) serum Analysis of mHOXA1 and mSST combination with CA19-9 showed to be useful to detect early stage of PDA. Suehiro et al. (2022)
[42]
cfDNA KRAS DF KRAS mutation analysis of DF collected after secretin administration showed high diagnostic accuracy for PDA. Yachida et al. (2025)
[43]
protein S100P DF The sensitivity and specificity of S100P protein expression in DF for diagnosing stages 0/IA/IB/IIA PDAC were 85% and 77%, respectively, with an AUC of 0.82. Ideno et al. (2020)
[45]
EV Glypican-1 (GPC1) serum GPC1-positive EVs, in combination with CD82 and CA19-9, have demonstrated high diagnostic accuracy for differentiating PDA from chronic pancreatitis (AUC 0.942). Xiao et al. (2020)
[46]
EV circRNAs (circ_0006220 and circ_0001666) plasma circ_0006220 and circ_0001666) were found to correlate with CA19-9 levels, tumor size, and lymph node metastasis; the combination of these two circRNAs yielded an AUC of 0.884 for PDA diagnosis. Hong et al. (2022)
[47]
EV 10 miRNAs plasma Ten miRNAs highly expressed in the body fluids of patients with PDA was selected using public databases; These miRNAs were identified and verified as EV-miRNA candidates for early detection. Makler et al. (2022)
[48]
EV 4 miRNAs plasma EV-miRNA panel comprising four miRNAs (miR-93-5p, miR-339-3p, and miR-425-5p/3p) achieved diagnostic accuracy comparable to or greater than CA19-9 (AUC 0.885). Makler et al. (2023)
[49]
EV molecular clustering of miRNAs plasma CT imaging features (radiomics) with the expression analysis of plasma EV-derived miRNAs (e.g., miR-1260b), improved the accuracy of differentiating malignant from benign pancreatic lesions to an AUC > 0.90. Xu et al. (2025)
[50]
EV miRNAs (e.g., miR-21, miR-10b, miR-451a) blood Systematic review demonstrated the utility of plasma and serum EV-derived miRNAs (e.g., miR-21, miR-10b, miR-451a) in the diagnosis of PDA. Patel et al. (2025)
[51]
EV HULC serum EV encapsulated HULC in serum was increased in serum derived from patients with PDA with AUC of 0.92. Takahashi et al. (2020)
[52]
DF; duodenal fluid,
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.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2026 MDPI (Basel, Switzerland) unless otherwise stated