
Diagnostic methods for pancreatic cancer and their clinical applications (Review)
- Authors:
- Published online on: May 27, 2025 https://doi.org/10.3892/ol.2025.15116
- Article Number: 370
-
Copyright: © Zhao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Pancreatic cancer (PC), particularly pancreatic ductal adenocarcinoma (PDAC), continues to be one of the most fatal cancers, marked by a poor prognosis and an elevated mortality rate. The American Cancer Society reports that PC is the 10th most common cancer in men and the 8th in women in terms of newly diagnosed cases. Furthermore, it is recognized as the fourth leading cause of mortality associated with cancer (1). Patients diagnosed with stage IV metastatic PC exhibit a 5-year survival rate of 3%. By contrast, patients presenting with stage I PC can expect a notably higher 5-year survival rate, reaching as much as 44% (2). Patients diagnosed with stage IA PC can attain a 5-year disease-free survival rate of 80% (3). This disease often presents without symptoms in its early stages, leading to advanced stage diagnoses where therapeutic options are limited and less effective (4). Consequently, timely identification and precise diagnosis are paramount to improving patient outcomes and survival rates.
The array of diagnostic techniques available for PC includes various modalities, each presenting distinct benefits and drawbacks. Imaging techniques such as ultrasound, computed tomography (CT), magnetic resonance imaging (MRI) and endoscopic ultrasound (EUS) are pivotal in the initial detection and staging of pancreatic lesions (5). These modalities provide critical anatomical and functional information that guides clinical decision-making. Biopsy techniques, such as percutaneous core-needle biopsy, EUS-guided fine-needle aspiration (FNA) and fine-needle biopsy, remain the gold standard for definitive diagnosis. These methods allow for histopathological and molecular analyses, providing essential information for personalized treatment strategies (6). However, challenges such as sample adequacy and procedural complications persist, highlighting the need for optimization and standardization (7). Tumor markers, particularly carbohydrate antigen 19-9 (CA19-9), play a significant role in the biochemical diagnosis of PC. Although CA19-9 is the most widely used serum marker, its specificity and sensitivity are suboptimal, necessitating the exploration of additional biomarkers to enhance diagnostic accuracy (8). Emerging diagnostic technologies, including liquid biopsy and advanced genomic and molecular techniques, are revolutionizing the field. Liquid biopsy, which analyzes indicators in the blood or other body fluids, offers a minimally invasive alternative for early detection and real-time monitoring of disease progression (9). Previous advances have identified several potential indicators, including circulating tumor cells (CTCs), circulating tumor (ct)DNA and exosomes, which hold promise for non-invasive diagnosis and monitoring (10). Additionally, genomic and molecular profiling of pancreatic tumors can uncover actionable mutations and guide targeted therapies, thereby enhancing precision medicine approaches (11). The metabolic changes within tumor cells have revealed that these cells release specific volatile organic compounds (VOCs), which could serve as potential indicators for the early identification of PC (12–14). Furthermore, the incorporation of nanomaterials into pre-existing technologies, such as sensors and biosensors, has resulted in notable improvements in both detection sensitivity and specificity (Fig. 1) (15).
The present review aims to discuss the latest research on imaging techniques, tumor markers, biopsy methods and novel diagnostic technologies, providing a holistic overview of their clinical applications and potential for integration into standard practice, offering insights into their efficacy, limitations and future directions.
Imaging techniques
Ultrasound, CT, MRI and EUS represent the cornerstone of non-invasive PC diagnostics. CT and MRI, with their superior anatomical resolution, have proven invaluable in pancreatic cancer staging and surgical planning. However, their sensitivity in detecting early-stage PC remains suboptimal. EUS, with its high-resolution images and potential for guided biopsies, has emerged as a critical tool (16). Artificial intelligence (AI) and machine learning (ML) algorithms are also being investigated to enhance diagnostic precision and optimize clinical workflows by analyzing complex data sets from imaging and molecular diagnostics (17).
Ultrasound examination
Transabdominal ultrasonography (TUS) examination is a widely used initial imaging modality for the diagnosis of PC due to its non-invasive nature, accessibility and cost-effectiveness. It employs high-frequency sound waves to produce images of the pancreas and surrounding structures. Despite its advantages, the sensitivity and specificity of ultrasound in detecting PC are relatively low, particularly for small lesions or pancreatic tail tumors (17). The accuracy of ultrasound can be significantly affected by obesity and bowel gas, which can obscure the pancreas. Previous studies have indicated that the presence of dilation and stenosis in the main pancreatic duct, pancreatic cyst formation, as well as localized adipose tissue alterations within the pancreas, as observed through imaging techniques, are distinctive features associated with the early stages of PDAC (18,19). Kanno et al (20) reported that the rate of tumor identification in stage I PC was 67.3% and the rate of main pancreatic duct dilation was 74.3% using TUS. Advancements in ultrasound technology, such as contrast-enhanced ultrasound (CEUS) and elastography, have improved diagnostic performance. CEUS enhances the visualization of vascular patterns within the tumor, aiding in the differentiation between benign and malignant lesions (21). CEUS has high diagnostic value in evaluating vascular invasion in patients with PDAC, especially invasion into the celiac artery and its branches (22). Elastography measures tissue stiffness, providing additional information that can help in the characterization of pancreatic masses (23).
EUS integrates endoscopy and ultrasound to provide high-resolution images of the pancreas and surrounding structures. EUS is valuable for detecting small pancreatic tumors and assessing local invasion and lymph node involvement (24). It allows for FNA of lesions, providing tissue samples for cytological examination and molecular analysis (25). The sensitivity of EUS in detecting PC is reported to be as high as 94%, with a specificity of 89% (26). Contrast-Enhanced Harmonic Imaging EUS (CEH EUS) has emerged as a superior tool for the early detection of smaller lesions, with studies showing 95.6% sensitivity compared to 82.7% for standard EUS (27). EUS is also valuable in guiding therapeutic interventions such as celiac plexus neurolysis for pain management in patients with PC (28). However, the accuracy of EUS can be operator-dependent, and the procedure is invasive, requiring sedation and carrying risks such as pancreatitis and infection.
CT and MRI
CT is considered the benchmark for the assessment and staging of PC. It offers comprehensive cross-sectional images of the pancreas, facilitating the evaluation of tumor size, location and involvement of adjacent structures. Multidetector CT with pancreatic protocol enhances the identification and characterization of pancreatic tumors (29). The sensitivity of CT in identifying PC is 76–92%, while the specificity is 85–95% (30). CT accurately detects PDAC at 98% for stage III but is less effective for stage I tumors (31). CT is also valuable in evaluating the resectability of the tumor by assessing vascular involvement, which is crucial for surgical planning. However, small tumors and those with isoattenuating characteristics may be missed on CT, necessitating the use of additional imaging modalities (32).
MRI is another essential imaging modality for the diagnosis and staging of PC. MRI offers enhanced contrast for soft tissues compared with CT, making it particularly useful for characterizing pancreatic lesions and detecting liver metastases (33). MRI techniques such as magnetic resonance cholangiopancreatography and diffusion-weighted imaging enhance the visualization of the pancreatic ductal system and tumor cellularity, respectively (34). Wiest et al (35) reported a sensitivity of 88–100%, a specificity of 63.4–94%, a positive predictive value (PPV) of 71.4–96.2% and a negative (N) PV of 68.5–100% for MRI. The sensitivity and specificity of MRI for detecting PC are comparable to those of CT, with some studies suggesting slightly higher sensitivity for small lesions (10,36). Additionally, MRI does not utilize ionizing radiation, which renders it a safer alternative for specific groups of patients. Positron emission tomography/CT (PET/CT) and positron emission tomography/MRI (PET/MRI) are not routinely used in the staging of patients with PDAC, may aid in detecting suspected pancreatic tumors (37).
AI-assisted diagnostic methods
In recent years, there has been a notable rise in the application of AI and ML within the healthcare sector (38). Similar to various other malignancies, screening, diagnosing and formulating treatment strategies for PDAC can be enhanced by AI models and ML algorithms. AI-driven analysis shows notable potential for enhancing the capabilities of imaging techniques in the early identification and characterization of PC (39). The present study consolidates current literature and a summary of a set of recent AI studies focused on the detection and diagnosis of PDAC, along with details regarding the models, datasets and evaluation metrics (40–63) (Table I).
Tissue biopsy
Percutaneous biopsy
Percutaneous biopsy, particularly CT-guided percutaneous FNA biopsy (FNAB), is widely used for diagnosing PC. This technique involves the insertion of a needle through the skin to obtain tissue samples from the pancreas, guided by imaging techniques such as CT or ultrasound. The accuracy of CT-guided percutaneous FNAB had a diagnostic accuracy rate of 95.1% in a study involving 84 patients with peritoneal lesions (64). The procedure is generally safe, with complications such as bleeding and ascite leakage occurring in a small percentage of cases (64). The high diagnostic yield and safety profile make percutaneous biopsy a valuable tool in the diagnostic arsenal for PC, especially when other less invasive methods fail to provide a definitive diagnosis.
Endoscopic-guided biopsy
EUS-guided FNAB (EUS-FNAB) has emerged as a preferred technique for obtaining tissue from pancreatic lesions. This technique integrates endoscopy and ultrasound to facilitate real-time imaging and precise needle placement. EUS-FNAB is especially advantageous for diagnosing PC, as it enables the sampling of lesions that are not easily accessible by percutaneous methods. Lee et al (65) have shown that EUS-FNAB has a high diagnostic accuracy, with sensitivity and specificity rates often >90%. Additionally, EUS-FNAB can be used to establish patient-derived PC organoids, which serve as valuable models for research and personalized treatment planning (32). Despite its high accuracy, there are instances where EUS-FNAB may fail to provide a definitive diagnosis, necessitating alternative methods such as serial pancreatic juice aspiration cytological examination (66).
FNAB
FNAB uses a thin needle to extract cells from a lesion for cytological examination. The procedure is often guided by ultrasound or CT to ensure accurate needle placement. FNAB has demonstrated efficacy in the diagnosis of PC, with study reporting high sensitivity (93%) and specificity rates (91%) (67). However, the diagnostic yield of FNAB can be influenced by factors such as the skill of the operator and the quality of the obtained sample. In cases where FNAB results are inconclusive, additional diagnostic methods or repeat biopsies may be necessary (68). The integration of molecular techniques, such as immunocytochemistry and genomic profiling, can enhance the diagnostic accuracy of FNAB and provide valuable information for personalized treatment planning (69).
Tissue biopsies, including percutaneous, endoscopic and FNA techniques, provide definitive histopathological diagnosis. However, the invasive nature and associated risks, such as pancreatitis and tumor seeding, limit their widespread use. Advances in biopsy techniques and the integration of molecular profiling could enhance diagnostic accuracy and guide personalized treatment strategies.
Tumor marker detection - CA19-9
CA19-9 is recognized as the most commonly utilized serum biomarker for PC. Despite its widespread use, CA19-9 has limitations, such as 80% sensitivity and 75% specificity, particularly in the early stages of PC (70). CA19-9 levels are elevated in ~80% of patients with advanced PC, but it can also be elevated in cholangitis, cirrhosis and other gastrointestinal malignancies, which complicates its diagnostic utility (10). Moreover, ~5-10% of patients with PC lack the Lewis antigen, which is necessary for CA19-9 expression, rendering this biomarker ineffective for these individuals (71). Its role in early detection remains limited, highlighting the necessity for supplementary biomarkers to enhance the precision of diagnostics. Joint detection of carcinoembryonic antigen (CEA), CA125, CA242 and CA19-9 may enhance the diagnostic efficiency for PC, increasing sensitivity to 90.4% and specificity to 93.8%, obviously higher than single detection of those markers in diagnosis of pancreatic cancer (72). Joint detection of MUC5AC and CA19-9 demonstrated enhanced performance and increased specificity in distinguishing PC from control groups, achieving an area under the curve (AUC) of 0.894, sensitivity of 0.738 and specificity of 0.886 (73). Combination of trefoil factors (TFFs) with CA19.9 emerged as a promising strategy for discriminating early-stage PC from benign controls (BC) (AUCTFF1 + TFF2 + TFF3 + CA19.9=0.93) as well as chronic pancreatitis (CP) (AUCTFF1 + TFF2 + TFF3 + CA19.9=0.93) (74). Combination of serum CA19-9 and serum glycoproteomics (IL.17E, B7.1 and DR6) can differentiate stage I PC from healthy controls (HCs; AUC=0.988; 100% sensitivity at 90% specificity) (75). However, these results still need to be further verified with large-scale, retrospective and prospective clinical studies. Given the limitations of CA19-9, research has focused on identifying additional biomarkers that could enhance the early identification and diagnosis of PC. The heterogeneity of PC and the lack of a universal biomarker underscore the necessity for a multimodal approach in biomarker-based diagnostics.
Emerging diagnostic technologies
Genomics and molecular biology techniques have revolutionized the field of cancer diagnostics, providing comprehensive understanding of the genetic and molecular characteristics of tumors. In PC, these techniques have facilitated the detection of specific genetic mutations and alterations that drive tumorigenesis. Next-generation sequencing has been instrumental in identifying actionable mutations in PC, guiding targeted therapy and personalized treatment approaches (76). Molecular biology techniques such as polymerase chain reaction (PCR) and digital droplet PCR (ddPCR) have also been employed to detect specific genetic alterations in PC. These techniques offer high sensitivity and specificity, allowing for the detection of low-abundance mutations in circulating free DNA (cfDNA) and exosomal DNA. ddPCR, in particular, has shown promise in detecting KRAS mutations in PC, providing valuable information for diagnosis and monitoring (77). The combination of CTCs, circulating free nucleic acids (cfNAs) and exosomes with advanced genomic analysis provides a comprehensive and non-invasive approach to PC diagnosis and monitoring (78).
Liquid biopsy is an innovative and minimally invasive diagnostic method that has shown promise in the early detection and monitoring of PC. This technique involves the analysis of CTCs, cfNAs and exosomes in bodily fluids. Studies have demonstrated the existence of CTCs associated with disease stage and prognosis (79,80). ctDNA, which refers to the diminutive segments of DNA that are emitted by neoplastic cells into the circulatory system, offers another promising avenue as it can provide information on genetic mutations, tumor burden and treatment responses (81). Exosomes, small extracellular vesicles containing proteins, lipids and nucleic acids, have also been recognized as promising biomarkers. Exosomal RNA and proteins can reflect the molecular characteristics of the tumor and have been shown to have diagnostic and prognostic value (82).
Diagnostic role of CTCs in PC
CTCs are malignant tumor cells present in the blood, which can either shed directly from primary tumor cells after undergoing epithelial-mesenchymal transition (EMT) or enter the bloodstream through the lymphatic system to reach secondary sites. They provide critical information regarding the genetic and phenotypic characteristics of the tumor, aiding in the diagnosis and monitoring of PC. Study has demonstrated that CTCs can be detected in patients with PC with varying sensitivity and specificity, making them a valuable tool for early diagnosis and prognosis (83). In a suitable environment, CTCs settle and proliferate, forming metastatic tumors, and are considered the ‘seeds’ of malignant tumor metastasis (84). CTCs are used as non-invasive assessment indicators for disease progression and prognosis in breast, colorectal and prostate cancer (85,86). In PC, patients with PDAC with positive CTCs have a poorer overall survival (OS) (HR, 1.23; 95% CI, 0.88–2.08; P<0.001) (87,88). Ankeny et al (89) found that the number of peripheral blood CTCs can serve as a biomarker for the diagnosis and staging of PC. However, there is still controversy regarding the use of CTCs for early detection of PDAC. Bidard et al (90) discovered that the detection rate of CTCs is low in the early stages of malignant tumors, with only 11% of patients with locally advanced PC having detectable levels of circulating CTCs. Tien et al (91) found that 68% of patients with PDAC had detectable CTCs in portal vein blood, while only 40% had detectable CTCs in peripheral blood, suggesting that portal vein blood may serve as a more effective alternative sample. However, currently, reliable separation and detection of CTCs remain technically challenging. On one hand, CTCs have a short half-life in peripheral blood (1–2.4 h) and low concentration (100,000–1,000,000/ml). On the other hand, CTC detection and enrichment rely on the utilization of highly specific biomarkers to attain the required levels of specificity and sensitivity. Furthermore, the high heterogeneity and plasticity of tumor cells complicate the selection of CTC marker detection (92).
Diagnostic role of cfNAs in PC
cfNAs, including ctDNA and ctRNA, are another component of liquid biopsy. These nucleic acids are released into the circulatory system from apoptotic and necrotic tumor cells. cfNAs can offer an extensive genetic characterization of the tumor, encompassing details such as mutations, variations in copy number and patterns of methylation. Previous advancements have improved the sensitivity and specificity of cfNA detection, making it a powerful tool for the early detection of PC (93). cfDNA released through tumor cell apoptosis, necrosis or active release is termed ctDNA, which accounts for a small portion of total cfDNA (94). ctDNA encompasses not only mutations that mirror those found in neoplastic cells but also demonstrates analogous epigenetic configurations, including DNA methylation, histone modifications and chromatin remodeling, which are consistent with gene expression and tumor characteristics (95). In the blood, ctDNA is swiftly eliminated from the bloodstream via the activity of endonucleases and exonucleases, in addition to renal excretion, exhibiting a half-life that varies between several minutes to 2 h (96). Therefore, it can reflect the actual condition of malignant tumors and the dynamic molecular changes during tumor development. Consequently, ctDNA has been used to understand the mutation status of malignant tumors, including PC (97). Additionally, ctDNA can also serve as a diagnostic method for diseases. Research has shown that the sensitivity and specificity of ctDNA for diagnosing PDAC are 65 and 75%, respectively, while the combined sensitivity and specificity of ctDNA, CA199 and CTCs increase to 78 and 91%, respectively. Combining ctDNA with CA199, CEA, hepatocyte growth factor and osteopontin can enhance the sensitivity to 64% and specificity to 99.5% of PDAC diagnosis, respectively (98). Zill et al (99) compared ctDNA samples from 26 patients with PC with tumor tissue sequences and assessed 54 gene mutations, finding that ctDNA mutations in genes including KRAS, TP53, APC, FBXW7 and SMAD could accurately detect PDAC. Li et al (100) have reported significant variability in the frequency of detected ctDNA mutations, such as KRAS in patients with PDAC ranging from 30 to 92%. Another option for detecting DNA mutations may be to examine variations in DNA methylation. Li et al (101) compared differentially methylated regions of cfDNA between patients with PC and HCs and developed a diagnostic prediction model incorporating MAPT, SIX3, MIR663, EPB41L3, FAM150A, TRIM73, LOC100128977 and LOC100130148, which serve as potential non-invasive diagnostic indicators for PC. Eissa et al (102) found that the ctDNA ADAMTS1 and BNC1 methylation could be used as diagnostic markers for early detection of PC with an AUC of 0.95 (sensitivity, 97.4%; specificity, 91.6%). However, ctDNA sequencing technology requires very high sensitivity and specificity to overcome the low concentration of ctDNA in the early stages of malignant tumors and the potential for false positives from cfDNA present in normal individuals (103).
miRNA is a non-coding RNA, consisting of ~21-25 nucleotides, that can affect the post-transcriptional expression of target genes. It is crucial for various essential biological functions, including development, proliferation and apoptosis. Studies have shown that miRNAs are present in saliva, serum, plasma and urine, with circulating miRNAs in the blood showing potential roles in the diagnosis of cancer. In the diagnosis of PC, Słotwiński and Slotwinska (104) found that plasma miR-16 and miR-196a combined with CA199 can better distinguish PC from HCs. In addition, Wang (105) discovered that serum miR-133a can differentiate PC from HC (AUC, 0.893; sensitivity, 90.6%; specificity, 87.2%). Furthermore, Wei et al (106) found that serum miR-1290 and miR-1246 combined with CA199 can effectively distinguish PC from HC (AUC, 0.97).
Due to the low abundance and fragility of circulating miRNAs in the bloodstream, they are easily degraded by RNase degradation in the bloodstream, leading to loss and/or damage during the extraction process. With advancements in research, it has been found that serum exosomes may serve as important carriers for circulating miRNAs. Exosomes can package miRNAs, protecting them from RNA enzyme digestion, and their high stability and ease of enrichment also address the challenges of enriching circulating miRNAs (107).
Diagnostic role of exosomes in PC
Exosomes are lipid-based vesicles released by cells into the extracellular environment, with a diameter of 40–160 nm and a phospholipid bilayer membrane structure. They encompass various bioactive components derived from the source cells, such as miRNA, mRNA, transcription factors, cytokines, growth factors and lipids. All cells can secrete exosomes, which can be found in various bodily fluids such as blood, urine, saliva, breast milk and bile. Initially, exosomes were considered to be a means for cells to release unwanted substances. However, subsequent research has revealed their significant role in facilitating intercellular communication and contributing to tumor advancement. The bioactive components contained within exosomes are closely related to the parent cells from which they originate, and thus, differences or specific expressions of some bioactive components in exosomes may make them potential biomarkers for the diagnosis of PC (108–145) (Table II).
Diagnostic role of exosomal RNA and DNA in PC
miRNAs in exosomes are relatively stable under various physicochemical conditions due to the protective membrane structure of exosomes, which shields them from ribonuclease digestion (146). Zhou et al (110) found that serum exosomal miR-122-5p and miR-193b-3p were upregulated in PC compared with HCs, while miR-221-3p was downregulated. Goto et al (111) compared 32 PC, 29 intraductal papillary mucinous neoplasm (IPMN) and 22 HC samples, and found that serum exosomal miR-191, miR-21 and miR-451a were upregulated in both PC and IPMN. Exosomal miRNAs could distinguish HCs from stage I and IIA PC, with miR-21 achieving a diagnostic accuracy of 80.8%. Additionally, exosomal miRNAs from other bodily fluids have shown potential diagnostic value for PC, such as salivary exosomal miR-1246 and miR-4644, which could serve as candidate biomarkers for diagnosing cholangiocarcinoma (AUC, 0.833). Yoshizawa et al (114) found that the proportion of miR-3940-5p to miR-8069 in urinary exosomes from patients with PDAC was elevated, achieving sensitivities and specificities of 93.0 and 78.4%, respectively, when combined with CA199 for diagnosing PDAC. To date, multiple studies have demonstrated that exosomal miRNAs, either individually or in conjunction, can serve as potential biomarkers for diagnosing PC (115,116).
Long noncoding RNAs (lncRNAs) are classified as noncoding RNAs that exceed a length of 200 nucleotides (147). Several studies have confirmed the abnormal expression of exosomal lncRNAs in PC (148). Takahashi et al (124) analyzed serum exosomal lncRNA-HULC in 20 cases of PDAC, 22 cases of IPMN and 21 HCs, finding that the expression of serum exosomal lncRNA-HULC in patients with PDAC was significantly increased compared with HCs and IPMN cases, showing good diagnostic performance (AUC, 0.920). Yu et al (126) studied 284 cases of PDAC, 100 cases of CP and 117 cases of HC, finding that a lncRNA group composed of FGA, KRT19, ITIH2, HIST1H2BK, MARCH2, CLDN1, MAL2 and TIMP1 showed high accuracy for the diagnosis of PDAC (AUC, 0.931). Circular RNA (circRNA) is also a noncoding RNA that has been discovered in recent years, and due to its closed-loop structure, exhibits higher stability compared with linear RNA (149). Li et al (127) conducted sequencing analysis of exosomal circRNA from the plasma of 8 patients with early-stage PDAC and 8 HCs, finding 155 circRNAs that were differentially expressed between PDAC and HC, which may be potential indicators for the early diagnosis of PDAC. In addition, small nucleolar RNA (snoRNA) is a non-coding RNA composed of ~60-300 nucleotides (150). Kitagawa et al (129) studied serum exosomes from 27 patients with stage I–II PDAC and 13 HCs, finding that SNORA74A and SNORA25l could serve as biomarkers for early detection of PDAC (AUC, 0.946 and 0.940, respectively). Kumar et al (132) sequenced serum exosomal mRNA and found that the expression of MMP8, TBX3, PDX1, CTSL and SIGLEC15 in serum exosomes obtained from PDAC samples was higher than that in HCs. Yang et al (131) found that the combination of circulating exosomal miR-409, CK18 mRNA, CD63 mRNA, circulating cfDNA concentration and CA19-9 had improved diagnostic efficacy for PC than CA19-9 testing alone (Table II).
The genomic mutations of DNA in exosomes can also serve as biomarkers for diagnosing PC (151). Allenson et al (134) found that plasma exosomal KRAS mutations in HCs and patients with early-stage, locally advanced and late-stage PDAC were 7.4, 66.7, 80.0 and 85.0%, respectively, with statistically significant differences. Furthermore, with the accumulation of exosomal analysis data, data mining identified 575 protein-coding genes, 26 RNA genes and 1 pseudogene directly related to PC (152). This exosome database established through pure bioinformatics methods serves as a valuable resource for the identification and confirmation of novel diagnostic combinations.
Diagnostic role of exosomal proteins and lipid components in PC
The diagnostic role of exosomal membrane proteins and the proteins contained within them in PC has also received considerable attention. GPC-1 is a proteoglycan located on the surface of cells that has been found to be significantly overexpressed in exosomes from prostate cancer cells and is considered a promising potential diagnostic marker in PDAC (153). Melo et al (137) extracted serum exosomes from HCs, benign pancreatic disease (BPD) and early to late-stage PDAC for mass spectrometry and nano-flow cytometry identification, finding that GPC-1+ exosomes could act as a marker for detecting early PC (AUC, 1). Buscai et al (154) discovered that combining CD63+GPC-1+ exosomes from peripheral blood and portal vein blood with CA19-9 improved the diagnostic efficacy for PC, and demonstrated a positive correlation between GPC-1− exosome levels and CTCs, which were associated with progression-free survival(PFS) and OS of patients. However, Lai et al (155) indicate that GPC-1 cannot diagnose PDAC. These studies confirm that GPC-1+ serum exosomes can act as potential diagnosis biomarkers for PC, but further research is needed for validation. Wei et al (141) found that serum exosomal Ephrin type-A receptor 2 (EphA2) could diagnose PC (AUC, 0.960). Additionally, exosomal proteins such as epidermal growth factor receptor (EGFR), alkaline phosphatase, placental like 2 (ALPPL2), cytoskeleton-associated protein 4 (CKAP4), cellular mesenchymal-epithelial transition factor (c-met), programmed death ligand 1 (PD-L1), epidermal growth factor receptor pathway substrate 8 (Eps8) and ALG-2-interacting protein X (ALIX) have also attracted the attention of researchers regarding their role in PC diagnosis (Table II) (140,143,144). Although further research is required for confirmation and there are differences in the types of proteins studied, the aforementioned studies indirectly suggest that exosomal membranes and contained proteins could serve as potential diagnosis biomarkers for PC.
Exosomes contain rich and complex lipid components such as cholesterol, sphingolipids and PS (156). Sharma et al (157) found that phosphatidylserine (PS) in plasma exosomes from patients with PC increased before histopathological confirmation of PC, suggesting it could be used for screening and ultra-early diagnosis of PC. Tao et al (158) applied liquid chromatography-mass spectrometry technology to analyze the lipid profiles of exosomes from the serum of patients with PC and HCs, identifying 37 differentially expressed lipid components. This suggests that these differentially expressed lipids may serve as potential biomarkers for diagnosing PC. However, there is currently limited research on exosomal lipids in the diagnosis of PC, making it difficult to evaluate their diagnostic efficacy.
The aforementioned research results indicate that circulating exosomal contents has potential value in the diagnosis and treatment of PC. However, due to the rich variety of exosomal contents, current studies are small-sample single-center studies, and there are differences in exosome extraction, content separation and sequencing methods. Additionally, body fluids are not uniform, and factors such as region, ethnicity and tumor staging can lead to differences in research results. Therefore, it is imperative to conduct large-sample multi-center studies after standardizing the technology, with the aim of finding suitable early diagnostic markers for PC.
In conclusion, emerging diagnostic technologies such as liquid biopsy and advanced genomic techniques hold significant promise for the early detection of PC. These methods offer a non-invasive, sensitive and specific approach to diagnosing PC. However, the integration of such sophisticated technologies into routine clinical practice poses challenges, including cost, accessibility and the need for specialized expertise. Further research and clinical validation are needed to fully realize the potential of these technologies in routine clinical practice.
Diagnostic role of VOCs in PC
VOCs are a diverse group of organic chemicals characterized by their elevated vapor pressure at ambient temperatures, which allows them to easily evaporate into the atmosphere. The unique metabolic activities of cancer cells can result in the production of specific VOCs that may serve as indicators of malignancy. The ability to detect these compounds in non-invasive biofluids such as breath, urine and saliva presents a significant advantage over traditional diagnostic methods, which often require invasive procedures such as biopsies. Research has shown that the presence and concentration of certain VOCs can correlate with the presence of pancreatic tumors, making them potential candidates for early diagnostic tools. Daulton et al (14) have shown that certain VOCs such as 2,6-dimethyl-octane and nonanal are significantly elevated in the urine of patients with PC compared with HCs, suggesting their potential as biomarkers for early detection (AUC, 0.85). Martínez-Moral et al (13) found that the serum VOC butoxymethylbenzene may be a suitable PC biomarker candidate (AUC, 0.98). Tiankanon et al (159) found that the VOCs, acetone dimers have the potential to be new biomarkers for PDAC detection (AUC, 0.91). A rapid, non-invasive and effective diagnostic method is particularly suitable for use in primary healthcare settings as a preliminary screening instrument, demonstrating significant patient acceptability and practicality. In the assessment of hepatobiliary conditions and PC, VOCs exhibited a sensitivity of 0.79 and a specificity of 0.81. These findings are notably encouraging and could offer a viable approach for the early identification and management of cancer (160).
Diagnostic role of nanomaterials in PC
The emergence of nanotechnology has paved the way for the development of sophisticated diagnostic tools, particularly through the implementation of nanomaterials. Nanomaterials can be engineered to target specific biomarkers linked to PC, facilitating more accurate detection techniques that could revolutionize the field of PC diagnostics (161). Caputo and Caracciolo (162) demonstrated that blood tests utilizing nanoparticles were capable of distinguishing patients with PDAC from healthy individuals through a comprehensive alteration in the nanoparticle-protein corona. Electrochemical biosensors utilizing nanomaterials have exhibited exceptional sensitivity for the detection of microRNAs and other biomarkers pertinent to PC, presenting opportunities for timely diagnosis (163).
Moccia et al (164) presented an economically viable paper-based electrochemical biosensor utilizing peptide nucleic acid for the identification of miRNA-492, a recognized biomarker associated with PDAC (164). The incorporation of nanomaterials into imaging technologies has markedly advanced the domain of medical diagnostics and treatment. The physicochemical attributes of nanomaterials, including their dimensions, morphology, surface chemistry and functionalization potential, contribute to enhanced imaging contrast, targeted delivery and heightened therapeutic effectiveness. For example, superparamagnetic iron oxide nanoparticles have been harnessed to target specific tumor markers, thereby enhancing the specificity of MRI for cancer identification (165). Gold nanoparticles have been utilized in photoacoustic imaging, wherein they absorb light and convert it into thermal energy, resulting in the generation of ultrasound signals that can be detected and visualized. This imaging technique has demonstrated potential in tumor imaging, offering deeper tissue penetration and superior resolution compared with conventional optical imaging methods (166). In summary, the future outlook for PC diagnostics appears promising with the emergence of nanomaterials. Ongoing research concentrating on developing efficient nanomaterial systems, combined with the integration of contemporary technologies and patient-centered methodologies, is crucial for advancing early detection and precision treatment.
Optimization of clinical pathways
Optimizing clinical pathways for PC diagnosis involves integrating multiple diagnostic modalities to enhance early detection and treatment planning. The combination of imaging techniques with tumor marker analysis and liquid biopsies can streamline the diagnostic process, reducing the time to diagnosis and improving patient outcomes. For example, a clinical pathway that incorporates initial imaging (CT or MRI) followed by EUS-FNA for suspicious lesions and concurrent CA19-9 testing, can provide a comprehensive diagnostic approach (10). Additionally, incorporating liquid biopsy techniques, such as ctDNA, exosome and VOCs analysis, can offer non-invasive options for monitoring disease progression and treatment response (167), and has become an essential instrument in improving the processes of screening, identifying, diagnosing, treating and monitoring PDAC (39). However, nanomaterials facilitate more accurate detection techniques, and thus implementing these integrated diagnostic pathways requires multidisciplinary collaboration and continuous evaluation to ensure they are tailored to individual patient needs and clinical settings (168). Through the integration of diverse diagnostic approaches and cutting-edge technologies, clinical pathways can be refined to enhance the rates of early detection and improve outcomes for patients with PC (10). An extensive diagnostic approach that combines imaging techniques, biomarker assessment and molecular characterization shows potential for enhancing early diagnosis and patient prognosis. Subsequent investigations should aim to concentrate on the validation of novel biomarkers, refinement of imaging techniques and the development of standardized protocols for liquid and molecular biopsies.
Conclusion
In the intricate landscape of PC diagnosis, it is evident that while notable strides have been made, there remains a critical need for advancements to enhance early identification and improve patient outcomes. The current diagnostic modalities, such as imaging techniques, tumor biomarker detection and tissue biopsies, each offer unique strengths but also present inherent limitations in sensitivity, specificity and clinical utility. Ultimately, the future of PC diagnosis is rooted in a multidisciplinary approach, leveraging advancements in technological innovations and enhanced comprehension of tumor biology. By addressing the current challenges and fostering collaborative research, progress can be made toward achieving early diagnosis and, consequently, improve prognoses for patients afflicted by PC.
Acknowledgements
Not applicable.
Funding
This work was supported by the Zhejiang Provincial Natural Science Foundation of China (grant no. LGC22H160012) and the Zhejiang Provincial Health Planning Commission Fund (grant no. 2023RC304).
Availability of data and materials
Not applicable.
Authors' contributions
Conceptualization, JZ; writing-original draft preparation, JW and YG; writing-review and editing, XH and LW. All authors read and approved the final manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Siegel RL, Miller KD, Wagle NS and Jemal A: Cancer statistics, 2023. CA Cancer J Clin. 73:17–48. 2023. View Article : Google Scholar : PubMed/NCBI | |
Huang C, Hecht EM, Soloff EV, Tiwari HA, Bhosale PR, Dasayam A, Galgano SJ, Kambadakone A, Kulkarni NM, Le O, et al: Imaging for early detection of pancreatic ductal adenocarcinoma: Updates and challenges in the implementation of screening and surveillance programs. AJR Am J Roentgenol. 223:e24311512024. View Article : Google Scholar : PubMed/NCBI | |
Blackford AL, Canto MI, Klein AP, Hruban RH and Goggins M: Recent trends in the incidence and survival of stage 1A pancreatic cancer: A surveillance, epidemiology, and end results analysis. J Natl Cancer Inst. 112:1162–1169. 2020. View Article : Google Scholar : PubMed/NCBI | |
Haberle L, Schramm M and Esposito I: Preoperative diagnostics of pancreatic neoplasms. Pathologe. 42:491–500. 2021.(In German). PubMed/NCBI | |
Bararia A, Chakraborty P, Roy P, Chattopadhay BK, Das A, Chatterjee A and Sikdar N: Emerging role of non-invasive and liquid biopsy biomarkers in pancreatic cancer. World J Gastroenterol. 29:2241–2260. 2023. View Article : Google Scholar : PubMed/NCBI | |
Schorr F and Essig MW: Early detection of pancreatic cancer - The role of endoscopic and transabdominal ultrasound. Z Gastroenterol. 59:1083–1090. 2021.(In German). PubMed/NCBI | |
Koziol-Bohatkiewicz P, Liberda-Matyja D and Wrobel TP: Fast cancer imaging in pancreatic biopsies using infrared imaging. Analyst. 149:1799–1806. 2024. View Article : Google Scholar : PubMed/NCBI | |
Zofia Rogowska A: Ultrasound-guided percutaneous core-needle biopsy of focal pancreatic lesions-practical aspectss. J Ultrason. 22:117–120. 2022. View Article : Google Scholar : PubMed/NCBI | |
Takahashi K, Takeda Y, Ono Y, Isomoto H and Mizukami Y: Current status of molecular diagnostic approaches using liquid biopsy. J Gastroenterol. 58:834–847. 2023. View Article : Google Scholar : PubMed/NCBI | |
Yang J, Xu R, Wang C, Qiu J, Ren B and You L: Early screening and diagnosis strategies of pancreatic cancer: A comprehensive review. Cancer Commun (Lond). 41:1257–1274. 2021. View Article : Google Scholar : PubMed/NCBI | |
Inchingolo R, Acquafredda F, Posa A, Nunes TF, Spiliopoulos S, Panzera F and Praticò CA: Endobiliary biopsy. World J Gastrointest Endosc. 14:291–301. 2022. View Article : Google Scholar : PubMed/NCBI | |
Navaneethan U, Spencer C, Zhu X, Vargo JJ, Grove D and Dweik RA: Volatile organic compounds in bile can distinguish pancreatic cancer from chronic pancreatitis: A prospective observational study. Endoscopy. 53:732–736. 2021. View Article : Google Scholar : PubMed/NCBI | |
Martinez-Moral MP, Tena MT, Martin-Carnicero A and Martinez A: Highly sensitive serum volatolomic biomarkers for pancreatic cancer diagnosis. Clin Chim Acta. 557:1178952024. View Article : Google Scholar : PubMed/NCBI | |
Daulton E, Wicaksono AN, Tiele A, Kocher HM, Debernardi S, Crnogorac-Jurcevic T and Covington JA: Volatile organic compounds (VOCs) for the non-invasive detection of pancreatic cancer from urine. Talanta. 221:1216042021. View Article : Google Scholar : PubMed/NCBI | |
Ma X, He C, Wang Y, Cao X, Jin Z, Ge Y, Cao Z, An M and Hao L: Mechanisms and applications of manganese-based nanomaterials in tumor diagnosis and therapy. Biomater Res. 29:01582025. View Article : Google Scholar : PubMed/NCBI | |
Hu X, Wang Z, Zhu Y, Li Z, Yan H, Zhao X and Wang Q: Advancements in molecular imaging for the diagnosis and treatment of pancreatic ductal adenocarcinoma. Nanoscale Adv. Apr 22–2025.(Epub ahead of print). View Article : Google Scholar | |
Sijithra PC, Santhi N and Ramasamy N: A review study on early detection of pancreatic ductal adenocarcinoma using artificial intelligence assisted diagnostic methods. Eur J Radiol. 166:1109722023. View Article : Google Scholar : PubMed/NCBI | |
Nakahodo J, Kikuyama M, Nojiri S, Chiba K, Yoshimoto K, Kamisawa T, Horiguchi SI and Honda G: Focal parenchymal atrophy of pancreas: An important sign of underlying high-grade pancreatic intraepithelial neoplasia without invasive carcinoma, i.e., carcinoma in situ. Pancreatology. 20:1689–1697. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yamao K, Takenaka M, Ishikawa R, Okamoto A, Yamazaki T, Nakai A, Omoto S, Kamata K, Minaga K, Matsumoto I, et al: Partial pancreatic parenchymal atrophy is a new specific finding to diagnose small pancreatic cancer (</=10 mm) including carcinoma in situ: Comparison with localized benign main pancreatic duct stenosis patients. Diagnostics (Basel). 10:4452020. View Article : Google Scholar : PubMed/NCBI | |
Kanno A, Masamune A, Hanada K, Maguchi H, Shimizu Y, Ueki T, Hasebe O, Ohtsuka T, Nakamura M, Takenaka M, et al: Multicenter study of early pancreatic cancer in Japan. Pancreatology. 18:61–67. 2018. View Article : Google Scholar : PubMed/NCBI | |
Kono Y, Oishi T, Ueda Y, Matsuoka E, Kamimura R, Tokiyoshi T, Okamoto T and Kashima S: A surgically resected case of lung metastases and sister Mary Joseph's Nodule 24 months after operation for pancreatic cancer. Gan To Kagaku Ryoho. 46:2354–2356. 2019.(In Japanese). PubMed/NCBI | |
Jia WY, Gui Y, Chen XQ, Tan L, Zhang J, Xiao MS, Chang XY, Dai MH, Guo JC, Cheng YJ, et al: Efficacy of color Doppler ultrasound and contrast-enhanced ultrasound in identifying vascular invasion in pancreatic ductal adenocarcinoma. Insights Imaging. 15:1812024. View Article : Google Scholar : PubMed/NCBI | |
Bonde A, Smith DA, Kikano E, Yoest JM, Tirumani SH and Ramaiya NH: Overview of serum and tissue markers in colorectal cancer: A primer for radiologists. Abdom Radiol (NY). 46:5521–5535. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhou D, Mu D, Cheng M, Dou Y, Zhang X, Feng Z, Qiu G, Yu H, Chen Y, Xu H, et al: Differences in lipidomics may be potential biomarkers for early diagnosis of pancreatic cancer. Acta Cir Bras. 35:e2020005082020. View Article : Google Scholar : PubMed/NCBI | |
Boyd LNC, Ali M, Comandatore A, Garajova I, Kam L, Puik JR, Fraga Rodrigues SM, Meijer LL, Le Large TYS, Besselink MG, et al: Prediction model for early-stage pancreatic cancer using routinely measured blood biomarkers. JAMA Netw Open. 6:e23311972023. View Article : Google Scholar : PubMed/NCBI | |
Guo W, Ying P, Ma R, Jing Z, Ma G, Long J, Li G and Liu Z: Liquid biopsy analysis of lipometabolic exosomes in pancreatic cancer. Cytokine Growth Factor Rev. 73:69–77. 2023. View Article : Google Scholar : PubMed/NCBI | |
Dahiya DS, Shah YR, Ali H, Chandan S, Gangwani MK, Canakis A, Ramai D, Hayat U, Pinnam BSM, Iqbal A, et al: Basic principles and role of endoscopic ultrasound in diagnosis and differentiation of pancreatic cancer from other pancreatic lesions: A comprehensive review of endoscopic ultrasound for pancreatic cancer. J Clin Med. 13:25992024. View Article : Google Scholar : PubMed/NCBI | |
Park JM, Mau CZ, Chen YC, Su YH, Chen HA, Huang SY, Chang JS and Chiu CF: A case-control study in Taiwanese cohort and meta-analysis of serum ferritin in pancreatic cancer. Sci Rep. 11:212422021. View Article : Google Scholar : PubMed/NCBI | |
Dias ESD and Chung V: Neoadjuvant treatment for pancreatic cancer: Controversies and advances. Cancer Treat Res Commun. 39:1008042024. View Article : Google Scholar : PubMed/NCBI | |
Roehnisch T, Martos-Contreras MC, Manoochehri M, Nogueira M, Bremm F, Dörrie J, Christoph J, Kunz M and Schönharting W: Individualized neoantigen peptide immunization of a metastatic pancreatic cancer patient: A case report of combined tumor and liquid biopsy. Front Immunol. 15:14147372024. View Article : Google Scholar : PubMed/NCBI | |
Singhi AD, Koay EJ, Chari ST and Maitra A: Early detection of pancreatic cancer: Opportunities and challenges. Gastroenterology. 156:2024–2040. 2019. View Article : Google Scholar : PubMed/NCBI | |
Ishikawa-Kakiya Y, Maruyama H, Kinoshita Y, Hayashi K, Yamamura M, Tanoue K, Nagami Y, Tanigawa T, Watanabe T and Fujiwara Y: The usefulness of serial pancreatic juice aspiration cytological examination for pancreatic cancer not diagnosed by EUS-FNAB. Clin J Gastroenterol. 13:1367–1372. 2020. View Article : Google Scholar : PubMed/NCBI | |
Chalfant H, Bonds M, Scott K, Condacse A, Dennahy IS, Martin WT, Little C, Edil BH, McNally LR and Jain A: Innovative imaging techniques used to evaluate borderline-resectable pancreatic adenocarcinoma. J Surg Res. 284:42–53. 2023. View Article : Google Scholar : PubMed/NCBI | |
Tantau A, Leucuta DC, Tantau M, Boţan E, Zaharie R, Mândruţiu A and Tomuleasa IC: Inflammation, tumoral markers and interleukin-17, −10, and −6 profiles in pancreatic adenocarcinoma and chronic pancreatitis. Dig Dis Sci. 66:3427–3438. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wiest NE, Moktan VP, Oman SP and Chirila RM: Screening for pancreatic cancer: A review for general clinicians. Rom J Intern Med. 58:119–128. 2020.PubMed/NCBI | |
Granata V, Fusco R, Setola SV, Galdiero R, Maggialetti N, Silvestro L, De Bellis M, Di Girolamo E, Grazzini G, Chiti G, et al: Risk assessment and pancreatic cancer: Diagnostic management and artificial intelligence. Cancers (Basel). 15:3512023. View Article : Google Scholar : PubMed/NCBI | |
Chu LC and Fishman EK: Pancreatic ductal adenocarcinoma staging: A narrative review of radiologic techniques and advances. Int J Surg. 110:6052–6063. 2024. View Article : Google Scholar : PubMed/NCBI | |
Koh DM, Papanikolaou N, Bick U, Illing R, Kahn CE Jr, Kalpathi-Cramer J, Matos C, Martí-Bonmatí L, Miles A, Mun SK, et al: Artificial intelligence and machine learning in cancer imaging. Commun Med (Lond). 2:1332022. View Article : Google Scholar : PubMed/NCBI | |
Mukund A, Afridi MA, Karolak A, Park MA, Permuth JB and Rasool G: Pancreatic ductal adenocarcinoma (PDAC): A review of recent advancements enabled by artificial intelligence. Cancers (Basel). 16:22402024. View Article : Google Scholar : PubMed/NCBI | |
Udristoiu AL, Cazacu IM, Gruionu LG, Gruionu G, Iacob AV, Burtea DE, Ungureanu BS, Costache MI, Constantin A, Popescu CF, et al: Real-time computer-aided diagnosis of focal pancreatic masses from endoscopic ultrasound imaging based on a hybrid convolutional and long short-term memory neural network model. PLoS One. 16:e02517012021. View Article : Google Scholar : PubMed/NCBI | |
Tong T, Gu J, Xu D, Song L, Zhao Q, Cheng F, Yuan Z, Tian S, Yang X, Tian J, et al: Deep learning radiomics based on contrast-enhanced ultrasound images for assisted diagnosis of pancreatic ductal adenocarcinoma and chronic pancreatitis. BMC Med. 20:742022. View Article : Google Scholar : PubMed/NCBI | |
Tonozuka R, Itoi T, Nagata N, Kojima H, Sofuni A, Tsuchiya T, Ishii K, Tanaka R, Nagakawa Y and Mukai S: Deep learning analysis for the detection of pancreatic cancer on endosonographic images: A pilot study. J Hepatobiliary Pancreat Sci. 28:95–104. 2021. View Article : Google Scholar : PubMed/NCBI | |
Marya NB, Powers PD, Chari ST, Gleeson FC, Leggett CL, Abu Dayyeh BK, Chandrasekhara V, Iyer PG, Majumder S, Pearson RK, et al: Utilisation of artificial intelligence for the development of an EUS-convolutional neural network model trained to enhance the diagnosis of autoimmune pancreatitis. Gut. 70:1335–1344. 2021. View Article : Google Scholar : PubMed/NCBI | |
Kuwahara T, Hara K, Mizuno N, Haba S, Okuno N, Kuraishi Y, Fumihara D, Yanaidani T, Ishikawa S, Yasuda T, et al: Artificial intelligence using deep learning analysis of endoscopic ultrasonography images for the differential diagnosis of pancreatic masses. Endoscopy. 55:140–149. 2023. View Article : Google Scholar : PubMed/NCBI | |
Ma H, Liu ZX, Zhang JJ, Wu FT, Xu CF, Shen Z, Yu CH and Li YM: Construction of a convolutional neural network classifier developed by computed tomography images for pancreatic cancer diagnosis. World J Gastroenterol. 26:5156–5168. 2020. View Article : Google Scholar : PubMed/NCBI | |
Liu SL, Li S, Guo YT, Zhou YP, Zhang ZD, Li S and Lu Y: Establishment and application of an artificial intelligence diagnosis system for pancreatic cancer with a faster region-based convolutional neural network. Chin Med J (Engl). 132:2795–2803. 2019. View Article : Google Scholar : PubMed/NCBI | |
Si K, Xue Y, Yu X, Zhu X, Li Q, Gong W, Liang T and Duan S: Fully end-to-end deep-learning-based diagnosis of pancreatic tumors. Theranostics. 11:1982–1990. 2021. View Article : Google Scholar : PubMed/NCBI | |
Qiu JJ, Yin J, Qian W, Liu JH, Huang ZX, Yu HP, Ji L and Zeng XX: A novel multiresolution-statistical texture analysis architecture: Radiomics-aided diagnosis of PDAC based on plain CT images. IEEE Trans Med Imaging. 40:12–25. 2021. View Article : Google Scholar : PubMed/NCBI | |
Qureshi TA, Gaddam S, Wachsman AM, Wang L, Azab L, Asadpour V, Chen W, Xie Y, Wu B, Pandol SJ and Li D: Predicting pancreatic ductal adenocarcinoma using artificial intelligence analysis of pre-diagnostic computed tomography images. Cancer Biomark. 33:211–217. 2022. View Article : Google Scholar : PubMed/NCBI | |
Ebrahimian S, Singh R, Netaji A, Madhusudhan KS, Homayounieh F, Primak A, Lades F, Saini S, Kalra MK and Sharma S: Characterization of benign and malignant pancreatic lesions with DECT quantitative metrics and radiomics. Acad Radiol. 29:705–713. 2022. View Article : Google Scholar : PubMed/NCBI | |
Chu LC, Park S, Kawamoto S, Fouladi DF, Shayesteh S, Zinreich ES, Graves JS, Horton KM, Hruban RH, Yuille AL, et al: Utility of CT radiomics features in differentiation of pancreatic ductal adenocarcinoma from normal pancreatic tissue. AJR Am J Roentgenol. 213:349–357. 2019. View Article : Google Scholar : PubMed/NCBI | |
Mukherjee S, Patra A, Khasawneh H, Korfiatis P, Rajamohan N, Suman G, Majumder S, Panda A, Johnson MP, Larson NB, et al: Radiomics-based machine-learning models can detect pancreatic cancer on prediagnostic computed tomography scans at a substantial lead time before clinical diagnosis. Gastroenterology. 163:1435–1446. e32022. View Article : Google Scholar : PubMed/NCBI | |
Li J, Liu F, Fang X, Cao K, Meng Y, Zhang H, Yu J, Feng X, Li Q, Liu Y, et al: CT radiomics features in differentiation of focal-type autoimmune pancreatitis from pancreatic ductal adenocarcinoma: A propensity score analysis. Acad Radiol. 29:358–366. 2022. View Article : Google Scholar : PubMed/NCBI | |
Ziegelmayer S, Kaissis G, Harder F, Jungmann F, Müller T, Makowski M and Braren R: Deep convolutional neural network-assisted feature extraction for diagnostic discrimination and feature visualization in pancreatic ductal adenocarcinoma (PDAC) versus autoimmune pancreatitis (AIP). J Clin Med. 9:40132020. View Article : Google Scholar : PubMed/NCBI | |
Cao K, Xia Y, Yao J, Han X, Lambert L, Zhang T, Tang W, Jin G, Jiang H, Fang X, et al: Large-scale pancreatic cancer detection via non-contrast CT and deep learning. Nat Med. 29:3033–3043. 2023. View Article : Google Scholar : PubMed/NCBI | |
Chen PT, Wu T, Wang P, Chang D, Liu KL, Wu MS, Roth HR, Lee PC, Liao WC and Wang W: pancreatic cancer detection on ct scans with deep learning: A nationwide population-based study. Radiology. 306:172–182. 2023. View Article : Google Scholar : PubMed/NCBI | |
Tayebi Arasteh S, Ziller A, Kuhl C, Makowski M, Nebelung S, Braren R, Rueckert D, Truhn D and Kaissis G: Preserving fairness and diagnostic accuracy in private large-scale AI models for medical imaging. Commun Med (Lond). 4:462024. View Article : Google Scholar : PubMed/NCBI | |
Liang Y, Schott D, Zhang Y, Wang Z, Nasief H, Paulson E, Hall W, Knechtges P, Erickson B and Li XA: Auto-segmentation of pancreatic tumor in multi-parametric MRI using deep convolutional neural networks. Radiother Oncol. 145:193–200. 2020. View Article : Google Scholar : PubMed/NCBI | |
Li J, Feng C, Lin X and Qian X: Utilizing GCN and meta-learning strategy in unsupervised domain adaptation for pancreatic cancer segmentation. IEEE J Biomed Health Inform. 26:79–89. 2022. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Chen Z, Li J, Zhang YD, Lin X and Qian X: Model-driven deep learning method for pancreatic cancer segmentation based on spiral-transformation. IEEE Trans Med Imaging. 41:75–87. 2022. View Article : Google Scholar : PubMed/NCBI | |
Li S, Jiang H, Wang Z, Zhang G and Yao YD: An effective computer aided diagnosis model for pancreas cancer on PET/CT images. Comput Methods Programs Biomed. 165:205–214. 2018. View Article : Google Scholar : PubMed/NCBI | |
Liu Z, Li M, Zuo C, Yang Z and Yang X, Ren S, Peng Y, Sun G, Shen J, Cheng C and Yang X: Radiomics model of dual-time 2-[(18)F]FDG PET/CT imaging to distinguish between pancreatic ductal adenocarcinoma and autoimmune pancreatitis. Eur Radiol. 31:6983–6991. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Cheng C, Liu Z, Wang L, Pan G, Sun G, Chang Y, Zuo C and Yang X: Radiomics analysis for the differentiation of autoimmune pancreatitis and pancreatic ductal adenocarcinoma in (18) F-FDG PET/CT. Med Phys. 46:4520–4530. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yu H, Zhang C, Liu S, Jiang G, Li S, Zhang L, Wang Y and Xu W: Clinical value of CT-guided percutaneous fine-needle aspiration biopsy for peritoneal lesions. BMC Med Imaging. 20:1222020. View Article : Google Scholar : PubMed/NCBI | |
Lee JH, Kim H, Lee SH, Ku JL, Chun JW, Seo HY, Kim SC, Paik WH, Ryu JK, Lee SK, et al: Establishment of patient-derived pancreatic cancer organoids from endoscopic ultrasound-guided fine-needle aspiration biopsies. Gut Liver. 16:625–636. 2022. View Article : Google Scholar : PubMed/NCBI | |
Masuda H, Kotecha K, Maitra R, Gill AJ, Mittal A and Samra JS: Clinical suspicion of pancreatic cancer despite negative endoscopic ultrasound-guided fine-needle aspiration biopsy. ANZ J Surg. 92:99–108. 2022. View Article : Google Scholar : PubMed/NCBI | |
Abe I and Lam AK: Fine-needle aspiration under guidance of ultrasound examination of thyroid lesions. Methods Mol Biol. 2534:29–37. 2022. View Article : Google Scholar : PubMed/NCBI | |
Smetanina SV, Slavnova EN, Smetanina OV, Golovin ST and Eremin NV: Features of differential cytological diagnosis of primary and metastatic liver carcinoma. Klin Lab Diagn. 66:364–370. 2021. View Article : Google Scholar : PubMed/NCBI | |
Taniuchi K, Ueno M, Yokose T, Sakaguchi M, Yoshioka R, Ogasawara M, Kosaki T, Naganuma S and Furihata M: Upregulation of PODXL and ITGB1 in pancreatic cancer tissues preoperatively obtained by EUS-FNAB correlates with unfavorable prognosis of postoperative pancreatic cancer patients. PLoS One. 17:e02651722022. View Article : Google Scholar : PubMed/NCBI | |
Mason J, Lundberg E, Jonsson P, Nyström H, Franklin O, Lundin C, Naredi P, Antti H, Sund M and Öhlund D: A cross-sectional and longitudinal analysis of pre-diagnostic blood plasma biomarkers for early detection of pancreatic cancer. Int J Mol Sci. 23:129692022. View Article : Google Scholar : PubMed/NCBI | |
Pietri E, Balsano R, Coriano M, Gelsomino F, Leonardi F, Bui S, Gnetti L, Valle RD and Garajová I: The implication of liquid biopsies to predict chemoresistance in pancreatic cancer. Cancer Drug Resist. 4:559–572. 2021.PubMed/NCBI | |
Gu YL, Lan C, Pei H, Yang SN, Liu YF and Xiao LL: Applicative Value of Serum CA19-9, CEA, CA125 and CA242 in diagnosis and prognosis for patients with pancreatic cancer treated by concurrent chemoradiotherapy. Asian Pac J Cancer Prev. 16:6569–6573. 2015. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Wang Y, Zhao T, Li Y, Tian L, Zhao J and Zhang J: Evaluation of serum MUC5AC in combination with CA19-9 for the diagnosis of pancreatic cancer. World J Surg Oncol. 18:312020. View Article : Google Scholar : PubMed/NCBI | |
Jahan R, Ganguly K, Smith LM, Atri P, Carmicheal J, Sheinin Y, Rachagani S, Natarajan G, Brand RE, Macha MA, et al: Trefoil factor(s) and CA19.9: A promising panel for early detection of pancreatic cancer. EBioMedicine. 42:375–385. 2019. View Article : Google Scholar : PubMed/NCBI | |
Aronsson L, Andersson R, Bauden M, Andersson B, Bygott T and Ansari D: High-density and targeted glycoproteomic profiling of serum proteins in pancreatic cancer and intraductal papillary mucinous neoplasm. Scand J Gastroenterol. 53:1597–1603. 2018. View Article : Google Scholar : PubMed/NCBI | |
Li X, Wang Q and Wang R: Roles of exosome genomic DNA in colorectal cancer. Front Pharmacol. 13:9232322022. View Article : Google Scholar : PubMed/NCBI | |
Wang ZY, Wang RX, Ding XQ, Zhang X, Pan XR and Tong JH: A protocol for cancer-related mutation detection on exosomal DNA in clinical application. Front Oncol. 10:5581062020. View Article : Google Scholar : PubMed/NCBI | |
Zhu Y, Zhang H, Chen N, Hao J, Jin H and Ma X: Diagnostic value of various liquid biopsy methods for pancreatic cancer: A systematic review and meta-analysis. Medicine (Baltimore). 99:e185812020. View Article : Google Scholar : PubMed/NCBI | |
Zhu Z, Zhang Y, Zhang W, Tang D, Zhang S, Wang L, Zou X, Ni Z, Zhang S, Lv Y and Xiang N: High-throughput enrichment of portal venous circulating tumor cells for highly sensitive diagnosis of CA19-9-negative pancreatic cancer patients using inertial microfluidics. Biosens Bioelectron. 259:1164112024. View Article : Google Scholar : PubMed/NCBI | |
Chen M, Liu H, Xiao Y, Liang R, Xu H, Hong B and Qian Y: Predictive biomarkers of pancreatic cancer metastasis: A comprehensive review. Clin Chim Acta. 569:1201762025. View Article : Google Scholar : PubMed/NCBI | |
Luo K, Wang X, Zhang X, Liu Z, Huang S and Li R: The value of circulating tumor cells in the prognosis and treatment of pancreatic cancer. Front Oncol. 12:9336452022. View Article : Google Scholar : PubMed/NCBI | |
Cui Y and Cao M: Liquid biopsy in bladder cancer. Methods Mol Biol. 2695:111–120. 2023. View Article : Google Scholar : PubMed/NCBI | |
Nagai M, Sho M, Akahori T, Nakagawa K and Nakamura K: Application of liquid biopsy for surgical management of pancreatic cancer. Ann Gastroenterol Surg. 4:216–223. 2020. View Article : Google Scholar : PubMed/NCBI | |
Gu X, Wei S and Lv X: Circulating tumor cells: From new biological insights to clinical practice. Signal Transduct Target Ther. 9:2262024. View Article : Google Scholar : PubMed/NCBI | |
Antonarakis ES, Lu C, Luber B, Wang H, Chen Y, Zhu Y, Silberstein JL, Taylor MN, Maughan BL, Denmeade SR, et al: Clinical significance of androgen receptor splice variant-7 mRNA detection in circulating tumor cells of men with metastatic castration-resistant prostate cancer treated with first- and second-line abiraterone and enzalutamide. J Clin Oncol. 35:2149–2156. 2017. View Article : Google Scholar : PubMed/NCBI | |
Silva ATF, Rodrigues CM, Ferreira ICC, Santos LLD, Santos DW, Araújo TG, Canto PPL, Paiva CE, Goulart LR and Maia YCP: A novel detection method of breast cancer through a simple panel of biomarkers. Int J Mol Sci. 23:119832022. View Article : Google Scholar : PubMed/NCBI | |
Earl J, Garcia-Nieto S, Martinez-Avila JC, Montans J, Sanjuanbenito A, Rodríguez-Garrote M, Lisa E, Mendía E, Lobo E, Malats N, et al: Circulating tumor cells (Ctc) and kras mutant circulating free Dna (cfdna) detection in peripheral blood as biomarkers in patients diagnosed with exocrine pancreatic cancer. BMC Cancer. 15:7972015. View Article : Google Scholar : PubMed/NCBI | |
Zhang ZH, Bao YW, Zhao YJ, Wang JQ, Guo JT and Sun SY: Circulating tumor cells as potential prognostic biomarkers for early-stage pancreatic cancer: A systematic review and meta-analysis. World J Clin Oncol. 14:504–517. 2023. View Article : Google Scholar : PubMed/NCBI | |
Ankeny JS, Court CM, Hou S, Li Q, Song M, Wu D, Chen JF, Lee T, Lin M, Sho S, et al: Circulating tumour cells as a biomarker for diagnosis and staging in pancreatic cancer. Br J Cancer. 114:1367–1375. 2016. View Article : Google Scholar : PubMed/NCBI | |
Bidard FC, Huguet F, Louvet C, Mineur L, Bouché O, Chibaudel B, Artru P, Desseigne F, Bachet JB, Mathiot C, et al: Circulating tumor cells in locally advanced pancreatic adenocarcinoma: the ancillary CirCe 07 study to the LAP 07 trial. Ann Oncol. 24:2057–2061. 2013. View Article : Google Scholar : PubMed/NCBI | |
Tien YW, Kuo HC, Ho BI, Chang MC, Chang YT, Cheng MF, Chen HL, Liang TY, Wang CF, Huang CY, et al: A high circulating tumor cell count in portal vein predicts liver metastasis from periampullary or pancreatic cancer: A high portal venous CTC count predicts liver metastases. Medicine (Baltimore). 95:e34072016. View Article : Google Scholar : PubMed/NCBI | |
Ahrens TD, Bang-Christensen SR, Jorgensen AM, Løppke C, Spliid CB, Sand NT, Clausen TM, Salanti A and Agerbæk MØ: The role of proteoglycans in cancer metastasis and circulating tumor cell analysis. Front Cell Dev Biol. 8:7492020. View Article : Google Scholar : PubMed/NCBI | |
Koo B, Jun E, Liu H, Kim EJ, Park YY, Lim SB, Kim SC and Shin Y: A biocomposite-based rapid sampling assay for circulating cell-free DNA in liquid biopsy samples from human cancers. Sci Rep. 10:149322020. View Article : Google Scholar : PubMed/NCBI | |
Bi F, Wang Q, Dong Q, Wang Y, Zhang L and Zhang J: Circulating tumor DNA in colorectal cancer: opportunities and challenges. Am J Transl Res. 12:1044–1055. 2020.PubMed/NCBI | |
Gai W and Sun K: Epigenetic biomarkers in cell-free DNA and applications in liquid biopsy. Genes (Basel). 10:322019. View Article : Google Scholar : PubMed/NCBI | |
Oliveira KCS, Ramos IB, Silva JMC, Barra WF, Riggins GJ, Palande V, Pinho CT, Frenkel-Morgenstern M, Santos SEB, Assumpcao PP, et al: Current perspectives on circulating tumor DNA, precision medicine, and personalized clinical management of cancer. Mol Cancer Res. 18:517–528. 2020. View Article : Google Scholar : PubMed/NCBI | |
Cescon DW, Bratman SV, Chan SM and Siu LL: Circulating tumor DNA and liquid biopsy in oncology. Nat Cancer. 1:276–290. 2020. View Article : Google Scholar : PubMed/NCBI | |
Topham JT, Renouf DJ and Schaeffer DF: Circulating tumor DNA: toward evolving the clinical paradigm of pancreatic ductal adenocarcinoma. Ther Adv Med Oncol. 15:175883592311576512023. View Article : Google Scholar : PubMed/NCBI | |
Zill OA, Greene C, Sebisanovic D, Siew LM, Leng J, Vu M, Hendifar AE, Wang Z, Atreya CE, Kelley RK, et al: Cell-Free DNA next-generation sequencing in pancreatobiliary carcinomas. Cancer Discov. 5:1040–1048. 2015. View Article : Google Scholar : PubMed/NCBI | |
Li H, Di Y, Li J, Jiang Y, He H, Yao L, Gu J, Lu J, Song J, Chen S, et al: Blood-based genomic profiling of circulating tumor DNA from patients with advanced pancreatic cancer and its value to guide clinical treatment. J Cancer. 11:4316–4323. 2020. View Article : Google Scholar : PubMed/NCBI | |
Li S, Wang L, Zhao Q, Wang Z, Lu S, Kang Y, Jin G and Tian J: Genome-wide analysis of cell-free DNA methylation profiling for the early diagnosis of pancreatic cancer. Front Genet. 11:5960782020. View Article : Google Scholar : PubMed/NCBI | |
Eissa MAL, Lerner L, Abdelfatah E, Shankar N, Canner JK, Hasan NM, Yaghoobi V, Huang B, Kerner Z, Takaesu F, et al: Promoter methylation of ADAMTS1 and BNC1 as potential biomarkers for early detection of pancreatic cancer in blood. Clin Epigenetics. 11:592019. View Article : Google Scholar : PubMed/NCBI | |
Hu Q, Chen L, Li K, Liu R, Sun L and Han T: Circulating tumor DNA: Current implementation issues and future challenges for clinical utility. Clin Chem Lab Med. 62:2094–2110. 2024. View Article : Google Scholar : PubMed/NCBI | |
Słotwiński R and Slotwinska SM: Diagnostic value of selected markers and apoptotic pathways for pancreatic cancer. Cent Eur J Immunol. 41:392–403. 2016. View Article : Google Scholar : PubMed/NCBI | |
Wang Z: Diagnostic performance for declined microRNA-133a in pancreatic cancer. J Cell Biochem. 121:3882–3886. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wei J, Yang L, Wu YN and Xu J: Serum miR-1290 and miR-1246 as Potential Diagnostic Biomarkers of Human Pancreatic Cancer. J Cancer. 11:1325–1333. 2020. View Article : Google Scholar : PubMed/NCBI | |
Kopcho S, McDew-White M, Naushad W, Mohan M and Okeoma CM: SIV Infection regulates compartmentalization of circulating blood plasma mirnas within extracellular vesicles (EVs) and extracellular condensates (ECs) and decreases EV-Associated miRNA-128. Viruses. 15:6222023. View Article : Google Scholar : PubMed/NCBI | |
Qin C, Li T, Lin C, Zhao B, Li Z, Zhao Y and Wang W: The systematic role of pancreatic cancer exosomes: Distant communication, liquid biopsy and future therapy. Cancer Cell Int. 24:2642024. View Article : Google Scholar : PubMed/NCBI | |
Zou X, Wei J, Huang Z, Zhou X, Lu Z, Zhu W and Miao Y: Identification of a six-miRNA panel in serum benefiting pancreatic cancer diagnosis. Cancer Med. 8:2810–2822. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhou X, Lu Z, Wang T, Huang Z, Zhu W and Miao Y: Plasma miRNAs in diagnosis and prognosis of pancreatic cancer: A miRNA expression analysis. Gene. 673:181–193. 2018. View Article : Google Scholar : PubMed/NCBI | |
Goto T, Fujiya M, Konishi H, Sasajima J, Fujibayashi S, Hayashi A, Utsumi T, Sato H, Iwama T, Ijiri M, et al: An elevated expression of serum exosomal microRNA-191, −21, −451a of pancreatic neoplasm is considered to be efficient diagnostic marker. BMC Cancer. 18:1162018. View Article : Google Scholar : PubMed/NCBI | |
Pu X, Ding G, Wu M, Zhou S, Jia S and Cao L: Elevated expression of exosomal microRNA-21 as a potential biomarker for the early diagnosis of pancreatic cancer using a tethered cationic lipoplex nanoparticle biochip. Oncol Lett. 19:2062–2070. 2020.PubMed/NCBI | |
Machida T, Tomofuji T, Maruyama T, Yoneda T, Ekuni D, Azuma T, Miyai H, Mizuno H, Kato H, Tsutsumi K, et al: miR-1246 and miR-4644 in salivary exosome as potential biomarkers for pancreatobiliary tract cancer. Oncol Rep. 36:2375–2381. 2016. View Article : Google Scholar : PubMed/NCBI | |
Yoshizawa N, Sugimoto K, Tameda M, Inagaki Y, Ikejiri M, Inoue H, Usui M, Ito M and Takei Y: miR-3940-5p/miR-8069 ratio in urine exosomes is a novel diagnostic biomarker for pancreatic ductal adenocarcinoma. Oncol Lett. 19:2677–2684. 2020.PubMed/NCBI | |
Shao H, Zhang Y, Yan J, Ban X, Fan X, Chang X, Lu Z, Wu Y, Zong L, Mo S, et al: Upregulated MicroRNA-483-3p is an early event in pancreatic ductal adenocarcinoma (PDAC) and as a powerful liquid biopsy biomarker in PDAC. Onco Targets Ther. 14:2163–2175. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang C, Wang J, Cui W, Liu Y, Zhou H, Wang Y, Chen X, Chen X and Wang Z: Serum Exosomal miRNA-1226 as potential biomarker of pancreatic ductal adenocarcinoma. Onco Targets Ther. 14:1441–1451. 2021. View Article : Google Scholar : PubMed/NCBI | |
Nakamura S, Sadakari Y, Ohtsuka T, Okayama T, Nakashima Y, Gotoh Y, Saeki K, Mori Y, Nakata K, Miyasaka Y, et al: Pancreatic juice exosomal MicroRNAs as biomarkers for detection of pancreatic ductal adenocarcinoma. Ann Surg Oncol. 26:2104–2111. 2019. View Article : Google Scholar : PubMed/NCBI | |
Chen J, Yao D, Chen W, Li Z, Guo Y, Zhu F and Hu X: Serum exosomal miR-451a acts as a candidate marker for pancreatic cancer. Int J Biol Markers. 37:74–80. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Wu J, Ye N, Li F, Zhan H, Chen S and Xu J: Plasma-derived exosome MiR-19b acts as a diagnostic marker for pancreatic cancer. Front Oncol. 11:7391112021. View Article : Google Scholar : PubMed/NCBI | |
Nakamura K, Zhu Z, Roy S, Jun E, Han H, Munoz RM, 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. 163:1252–1266. e22022. View Article : Google Scholar : PubMed/NCBI | |
Guo S, Qin H, Liu K, Wang H, Bai S, Liu S, Shao Z, Zhang Y, Song B, Xu X, et al: Blood small extracellular vesicles derived miRNAs to differentiate pancreatic ductal adenocarcinoma from chronic pancreatitis. Clin Transl Med. 11:e5202021. View Article : Google Scholar : PubMed/NCBI | |
Chen J, Zhang X, Zhang G, Zhu F and Liu W: Serum-derived exosomal miR-7977 combined with miR-451a as a potential biomarker for pancreatic ductal adenocarcinoma. BMC Cancer. 25:2952025. View Article : Google Scholar : PubMed/NCBI | |
Taniguchi T, Ideno N, Araki T, Miura S, Yamamoto M, Nakafusa T, Higashijima N, Yamamoto T, Tamura K, Nakamura S, et al: MicroRNA-20a in extracellular vesicles derived from duodenal fluid is a possible biomarker for pancreatic ductal adenocarcinoma. DEN Open. 4:e3332024. View Article : Google Scholar : PubMed/NCBI | |
Takahashi K, Ota Y, Kogure T, Suzuki Y, Iwamoto H, Yamakita K, Kitano Y, Fujii S, Haneda M, Patel T and Ota T: Circulating extracellular vesicle-encapsulated HULC is a potential biomarker for human pancreatic cancer. Cancer Sci. 111:98–111. 2020. View Article : Google Scholar : PubMed/NCBI | |
Kumar SR, Kimchi ET, Manjunath Y, Gajagowni S, Stuckel AJ and Kaifi JT: Author Correction: RNA cargos in extracellular vesicles derived from blood serum in pancreas associated conditions. Sci Rep. 10:99812020. View Article : Google Scholar : PubMed/NCBI | |
Yu S, Li Y, Liao Z, Wang Z, Li Y, Qian L, Zhao J, Zong H, Kang B, Zou WB, et al: Plasma extracellular vesicle long RNA profiling identifies a diagnostic signature for the detection of pancreatic ductal adenocarcinoma. Gut. 69:540–550. 2020. View Article : Google Scholar : PubMed/NCBI | |
Li J, Li Z, Jiang P, Peng M, Zhang X, Chen K, Liu H, Bi H, Liu X and Li X: Circular RNA IARS (circ-IARS) secreted by pancreatic cancer cells and located within exosomes regulates endothelial monolayer permeability to promote tumor metastasis. J Exp Clin Cancer Res. 37:1772018. View Article : Google Scholar : PubMed/NCBI | |
Hong L, Xu L, Jin L, Xu K, Tang W, Zhu Y, Qiu X and Wang J: Exosomal circular RNA hsa_circ_0006220, and hsa_circ_0001666 as biomarkers in the diagnosis of pancreatic cancer. J Clin Lab Anal. 36:e244472022. View Article : Google Scholar : PubMed/NCBI | |
Kitagawa T, Taniuchi K, Tsuboi M, Sakaguchi M, Kohsaki T, Okabayashi T and Saibara T: Circulating pancreatic cancer exosomal RNAs for detection of pancreatic cancer. Mol Oncol. 13:212–227. 2019. View Article : Google Scholar : PubMed/NCBI | |
Hu J, Sheng Y, Kwak KJ, Shi J, Yu B and Lee LJ: A signal-amplifiable biochip quantifies extracellular vesicle-associated RNAs for early cancer detection. Nat Commun. 8:16832017. View Article : Google Scholar : PubMed/NCBI | |
Yang Z, LaRiviere MJ, Ko J, Till JE, Christensen T, Yee SS, Black TA, Tien K, Lin A, Shen H, et al: A multianalyte panel consisting of extracellular vesicle miRNAs and mRNAs, cfDNA, and CA19-9 shows utility for diagnosis and staging of pancreatic ductal adenocarcinoma. Clin Cancer Res. 26:3248–3258. 2020. View Article : Google Scholar : PubMed/NCBI | |
Kumar SR, Kimchi ET, Manjunath Y, Gajagowni S, Stuckel AJ and Kaifi JT: RNA cargos in extracellular vesicles derived from blood serum in pancreas associated conditions. Sci Rep. 10:28002020. View Article : Google Scholar : PubMed/NCBI | |
Wang F, Wang C, Chen S, Wei C, Ji J, Liu Y, Liang L, Chen Y, Li X, Zhao L, et al: Identification of blood-derived exosomal tumor RNA signatures as noninvasive diagnostic biomarkers for multi-cancer: A multi-phase, multi-center study. Mol Cancer. 24:602025. View Article : Google Scholar : PubMed/NCBI | |
Allenson K, Castillo J, San Lucas FA, Scelo G, Kim DU, Bernard V, Davis G, Kumar T, Katz M, Overman MJ, et al: High prevalence of mutant KRAS in circulating exosome-derived DNA from early-stage pancreatic cancer patients. Ann Oncol. 28:741–747. 2017. View Article : Google Scholar : PubMed/NCBI | |
Castillo J, Bernard V, San Lucas FA, Allenson K, Capello M, Kim DU, Gascoyne P, Mulu FC, Stephens BM, Huang J, et al: Surfaceome profiling enables isolation of cancer-specific exosomal cargo in liquid biopsies from pancreatic cancer patients. Ann Oncol. 29:223–229. 2018. View Article : Google Scholar : PubMed/NCBI | |
Jin H, Liu P, Wu Y, Meng X, Wu M, Han J and Tan X: Exosomal zinc transporter ZIP4 promotes cancer growth and is a novel diagnostic biomarker for pancreatic cancer. Cancer Sci. 109:2946–2956. 2018. View Article : Google Scholar : PubMed/NCBI | |
Melo SA, Luecke LB, Kahlert C, Fernandez AF, Gammon ST, Kaye J, LeBleu VS, Mittendorf EA, Weitz J, Rahbari N, et al: Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature. 523:177–182. 2015. View Article : Google Scholar : PubMed/NCBI | |
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. 18:300–310. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yang KS, Im H, Hong S, Pergolini I, Del Castillo AF, Wang R, Clardy S, Huang CH, Pille C, Ferrone S, et al: Multiparametric plasma EV profiling facilitates diagnosis of pancreatic malignancy. Sci Transl Med. 9:eaal32262017. View Article : Google Scholar : PubMed/NCBI | |
Yang J, Zhang Y, Gao X, Yuan Y, Zhao J, Zhou S, Wang H, Wang L, Xu G, Li X, et al: Plasma-derived exosomal ALIX as a novel biomarker for diagnosis and classification of pancreatic cancer. Front Oncol. 11:6283462021. View Article : Google Scholar : PubMed/NCBI | |
Wei Q, Zhang J, Li Z, Wei L and Ren L: Serum Exo-EphA2 as a potential diagnostic biomarker for pancreatic cancer. Pancreas. 49:1213–1219. 2020. View Article : Google Scholar : PubMed/NCBI | |
Liang K, Liu F, Fan J, Sun D, Liu C, Lyon CJ, Bernard DW, Li Y, Yokoi K, Katz MH, et al: Nanoplasmonic quantification of tumor-derived extracellular vesicles in plasma microsamples for diagnosis and treatment monitoring. Nat Biomed Eng. 1:002112017 View Article : Google Scholar : PubMed/NCBI | |
Shin HS, Jung SB, Park S, Dua P and Lee DK: ALPPL2 Is a potential diagnostic biomarker for pancreatic cancer-derived extracellular vesicles. Mol Ther Methods Clin Dev. 15:204–210. 2019. View Article : Google Scholar : PubMed/NCBI | |
Lux A, Kahlert C, Grutzmann R and Pilarsky C: c-Met and PD-L1 on circulating exosomes as diagnostic and prognostic markers for pancreatic cancer. Int J Mol Sci. 20:33052019. View Article : Google Scholar : PubMed/NCBI | |
David P, Kouhestani D, Hansen FJ, Paul S, Czubayko F, Karabiber A, Weisel N, Klösch B, Merkel S, Ole-Baur J, et al: Exosomal CD40, CD25, and Serum CA19-9 as combinatory novel liquid biopsy biomarker for the diagnosis and prognosis of patients with pancreatic ductal adenocarcinoma. Int J Mol Sci. 26:15002025. View Article : Google Scholar : PubMed/NCBI | |
Fan Z, Yu J, Lin J, Liu Y and Liao Y: Exosome-specific tumor diagnosis via biomedical analysis of exosome-containing microRNA biomarkers. Analyst. 144:5856–5865. 2019. View Article : Google Scholar : PubMed/NCBI | |
Long Y, Wang X, Youmans DT and Cech TR: How do lncRNAs regulate transcription? Sci Adv. 3:eaao21102017. View Article : Google Scholar : PubMed/NCBI | |
Pandya G, Kirtonia A, Sethi G, Pandey AK and Garg M: The implication of long non-coding RNAs in the diagnosis, pathogenesis and drug resistance of pancreatic ductal adenocarcinoma and their possible therapeutic potential. Biochim Biophys Acta Rev Cancer. 1874:1884232020. View Article : Google Scholar : PubMed/NCBI | |
Lin H, Yu J, Gu X, Ge S and Fan X: Novel insights into exosomal circular RNAs: Redefining intercellular communication in cancer biology. Clin Transl Med. 11:e6362021. View Article : Google Scholar : PubMed/NCBI | |
Chen Q and Zhou T: Emerging functional principles of tRNA-derived small RNAs and other regulatory small RNAs. J Biol Chem. 299:1052252023. View Article : Google Scholar : PubMed/NCBI | |
Guo S, Wang X, Shan D, Xiao Y, Ju L, Zhang Y, Wang G and Qian K: The detection, biological function, and liquid biopsy application of extracellular vesicle-associated DNA. Biomark Res. 12:1232024. View Article : Google Scholar : PubMed/NCBI | |
Makler A and Narayanan R: Mining exosomal genes for pancreatic cancer targets. Cancer Genomics Proteomics. 14:161–172. 2017. View Article : Google Scholar : PubMed/NCBI | |
Li H, Chiang CL, Kwak KJ, Wang X, Doddi S, Ramanathan LV, Cho SM, Hou YC, Cheng TS, Mo X, et al: Extracellular vesicular analysis of glypican 1 mRNA and protein for pancreatic cancer diagnosis and prognosis. Adv Sci (Weinh). 11:e23063732024. View Article : Google Scholar : PubMed/NCBI | |
Buscail E, Chauvet A, Quincy P, Degrandi O, Buscail C, Lamrissi I, Moranvillier I, Caumont C, Verdon S, Brisson A, et al: CD63-GPC1-positive exosomes coupled with CA19-9 offer good diagnostic potential for resectable pancreatic ductal adenocarcinoma. Transl Oncol. 12:1395–1403. 2019. View Article : Google Scholar : PubMed/NCBI | |
Lai X, Wang M, McElyea SD, Sherman S, House M and Korc M: A microRNA signature in circulating exosomes is superior to exosomal glypican-1 levels for diagnosing pancreatic cancer. Cancer Lett. 393:86–93. 2017. View Article : Google Scholar : PubMed/NCBI | |
Skotland T, Hessvik NP, Sandvig K and Llorente A: Exosomal lipid composition and the role of ether lipids and phosphoinositides in exosome biology. J Lipid Res. 60:9–18. 2019. View Article : Google Scholar : PubMed/NCBI | |
Sharma R, Huang X, Brekken RA and Schroit AJ: Detection of phosphatidylserine-positive exosomes for the diagnosis of early-stage malignancies. Br J Cancer. 117:545–552. 2017. View Article : Google Scholar : PubMed/NCBI | |
Tao L, Zhou J, Yuan C, Zhang L, Li D, Si D, Xiu D and Zhong L: Metabolomics identifies serum and exosomes metabolite markers of pancreatic cancer. Metabolomics. 15:862019. View Article : Google Scholar : PubMed/NCBI | |
Tiankanon K, Pungpipattrakul N, Sukaram T, Chaiteerakij R and Rerknimitr R: Identification of breath volatile organic compounds to distinguish pancreatic adenocarcinoma, pancreatic cystic neoplasm, and patients without pancreatic lesions. World J Gastrointest Oncol. 16:894–906. 2024. View Article : Google Scholar : PubMed/NCBI | |
Pelling M, Chandrapalan S, West E and Arasaradnam RP: A systematic review and meta-analysis: Volatile organic compound analysis in the detection of hepatobiliary and pancreatic cancers. Cancers (Basel). 15:23082023. View Article : Google Scholar : PubMed/NCBI | |
Nagarajan Y, Chandrasekaran N and Deepa Parvathi V: Functionalized nanomaterials in pancreatic cancer theranostics and molecular imaging. ChemistryOpen. 14:e2024002322025. View Article : Google Scholar : PubMed/NCBI | |
Caputo D and Caracciolo G: Nanoparticle-enabled blood tests for early detection of pancreatic ductal adenocarcinoma. Cancer Lett. 470:191–196. 2020. View Article : Google Scholar : PubMed/NCBI | |
Yin X, He Z, Ge W and Zhao Z: Application of aptamer functionalized nanomaterials in targeting therapeutics of typical tumors. Front Bioeng Biotechnol. 11:10929012023. View Article : Google Scholar : PubMed/NCBI | |
Moccia M, Caratelli V, Cinti S, Pede B, Avitabile C, Saviano M, Imbriani AL, Moscone D and Arduini F: Paper-based electrochemical peptide nucleic acid (PNA) biosensor for detection of miRNA-492: A pancreatic ductal adenocarcinoma biomarker. Biosens Bioelectron. 165:1123712020. View Article : Google Scholar : PubMed/NCBI | |
Carigga Gutierrez NM, Clainche TL, Bulin AL, Leo S, Kadri M, Abdelhamid AGA, Pujol-Solé N, Obaid G, Hograindleur MA, Gardette V, et al: Engineering radiocatalytic nanoliposomes with hydrophobic gold nanoclusters for radiotherapy enhancement. Adv Mater. 36:e24046052024. View Article : Google Scholar : PubMed/NCBI | |
Phua VJX, Yang CT, Xia B, Yan SX, Liu J, Aw SE, He T and Ng DCE: Nanomaterial probes for nuclear imaging. Nanomaterials (Basel). 12:5822022. View Article : Google Scholar : PubMed/NCBI | |
Kaczor-Urbanowicz KE, Cheng J, King JC, Sedarat A, Pandol SJ, Farrell JJ, Wong DTW and Kim Y: Reviews on current liquid biopsy for detection and management of pancreatic cancers. Pancreas. 49:1141–1152. 2020. View Article : Google Scholar : PubMed/NCBI | |
Qiao Z, Ge J, He W, Xu X and He J: Artificial intelligence algorithm-based computerized tomography image features combined with serum tumor markers for diagnosis of pancreatic cancer. Comput Math Methods Med. 2022:89794042022. View Article : Google Scholar : PubMed/NCBI |