Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Pancreatology ; 2024 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-39299885

RESUMEN

RATIONALE AND OBJECTIVES: To investigate whether computed tomography features can differentiate pancreatoblastoma (PB) from solid pseudopapillary tumor (SPN) in children. MATERIALS AND METHODS: Clinical and imaging data of 18 cases of PB and 61 cases of SPN confirmed by surgery or biopsy were retrospectively analyzed. All enrolled patients underwent 3 phases (non-contrast, arterial, and portal venous phases) of CT scanning. Qualitative CT analysis (location, margin, solid/cystic component proportion, calcification, hemorrhage, peritumoral vascularity, bile duct dilatation, pancreatic duct dilatation, pancreatic atrophy, vascular invasion, peripancreatic invasion, and distant metastases) and quantitative analysis (maximum tumor diameter, interface between tumor and parenchyma [delta], arterial enhancement ratio [AER], and portal enhancement ratio [PER]) were performed. The general CT morphologic features, age and tumor markers were compared also compared between the groups. Univariate analysis and the F test were conducted to identify features of PB. Then logistic Regression classifier was trained using the top five features with the highest F-value. Moreover, we used 5-fold cross-validation techniques for the validation of our model. RESULTS: PB exhibited a significantly higher frequency of location in the body/tail, larger tumor size, poorly defined margins, calcification, peritumoral vascularity, pancreatic atrophy, and less hemorrhage. In addition, PB had higher AER, PER and lower delta relative to SPN (p < 0.05). PB presented a younger age and higher levels of AFP. Results of the F test indicated that AFP, AER, Age, calcification and pancreatic atrophy were the top five features included in the model that could differentiate pediatric PB from SPN. The combined model of CT and clinical features performed well in differentiating PB from SPN, with an AUC of 0.981 in the training cohort and 0.953 in the validation cohort. CONCLUSIONS: AFP, AER, age, calcification and pancreatic atrophy are robust CT and clinical features for differentiating pediatric PB from SPN. A combination of qualitative and quantitative CT features may provide good diagnostic accuracy in differentiating PB from SPN in children.

2.
World J Gastrointest Surg ; 16(7): 1986-2002, 2024 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-39087130

RESUMEN

A pancreatic pseudocyst is defined as an encapsulated fluid collection with a well-defined inflammatory wall with minimal or no necrosis. The diagnosis cannot be made prior to 4 wk after the onset of pancreatitis. The clinical presentation is often nonspecific, with abdominal pain being the most common symptom. If a diagnosis is suspected, contrast-enhanced computed tomography and/or magnetic resonance imaging are performed to confirm the diagnosis and assess the characteristics of the pseudocyst. Endoscopic ultrasound with cyst fluid analysis can be performed in cases of diagnostic uncertainty. Pseudocyst of the pancreas can lead to complications such as hemorrhage, infection, and rupture. The management of pancreatic pseudocysts depends on the presence of symptoms and the development of complications, such as biliary or gastric outlet obstruction. Management options include endoscopic or surgical drainage. The aim of this review was to summarize the current literature on pancreatic pseudocysts and discuss the evolution of the definitions, diagnosis, and management of this condition.

3.
Res Diagn Interv Imaging ; 9: 100038, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-39076579

RESUMEN

Objective: The objective of this study was to evaluate the clinical feasibility of deep learning reconstruction-accelerated thin-slice single-breath-hold half-Fourier single-shot turbo spin echo imaging (HASTEDL) for detecting pancreatic lesions, in comparison with two conventional T2-weighted imaging sequences: compressed-sensing HASTE (HASTECS) and BLADE. Methods: From March 2022 to January 2023, a total of 63 patients with suspected pancreatic-related disease underwent the HASTEDL, HASTECS, and BLADE sequences were enrolled in this retrospectively study. The acquisition time, the pancreatic lesion conspicuity (LCP), respiratory motion artifact (RMA), main pancreatic duct conspicuity (MPDC), overall image quality (OIQ), signal-to-noise ratio (SNR), and contrast-noise-ratio (CNR) of the pancreatic lesions were compared among the three sequences by two readers. Results: The acquisition time of both HASTEDL and HASTECS was 16 s, which was significantly shorter than that of 102 s for BLADE. In terms of qualitative parameters, Reader 1 and Reader 2 assigned significantly higher scores to the LCP, RMA, MPDC, and OIQ for HASTEDL compared to HASTECS and BLADE sequences; As for the quantitative parameters, the SNR values of the pancreatic head, body, tail, and lesions, the CNR of the pancreatic lesion measured by the two readers were also significantly higher for HASTEDL than for HASTECS and BLADE sequences. Conclusions: Compared to conventional T2WI sequences (HASTECS and BLADE), deep-learning reconstructed HASTE enables thin slice and single-breath-hold acquisition with clinical acceptable image quality for detection of pancreatic lesions.

4.
Endosc Ultrasound ; 13(2): 83-88, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38947744

RESUMEN

Background and Objectives: Pancreatic cancer (PC) is the third cause of cancer-related deaths. Early detection and interception of premalignant pancreatic lesions represent a promising strategy to improve outcomes. We evaluated risk factors of focal pancreatic lesions (FPLs) in asymptomatic individuals at hereditary high risk for PC. Methods: This is an observational single-institution cohort study conducted over a period of 5 years. Surveillance was performed through imaging studies (EUS or magnetic resonance imaging/magnetic resonance cholangiopancreatography) and serum biomarkers. We collected demographic characteristics and used univariate and multivariate logistic regression models to evaluate associations between potential risk factors and odd ratios (ORs) for FPL development. Results: A total of 205 patients completed baseline screening. Patients were followed up to 53 months. We detected FPL in 37 patients (18%) at baseline; 2 patients had lesions progression during follow-up period, 1 of them to PC. Furthermore, 13 patients developed new FPLs during the follow-up period. Univariate and multivariate analyses revealed that new-onset diabetes (NOD) is strongly associated with the presence of FPL (OR, 10.94 [95% confidence interval, 3.01-51.79; P < 0.001]; OR, 9.98 [95% confidence interval, 2.15-46.33; P = 0.003]). Follow-up data analysis revealed that NOD is also predictive of lesions progression or development of new lesions during screening (26.7% vs. 2.6%; P = 0.005). Conclusions: In a PC high-risk cohort, NOD is significantly associated with presence of FPL at baseline and predictive of lesions progression or new lesions during surveillance.

5.
Endosc Ultrasound ; 13(2): 65-75, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38947752

RESUMEN

Artificial intelligence (AI) is an epoch-making technology, among which the 2 most advanced parts are machine learning and deep learning algorithms that have been further developed by machine learning, and it has been partially applied to assist EUS diagnosis. AI-assisted EUS diagnosis has been reported to have great value in the diagnosis of pancreatic tumors and chronic pancreatitis, gastrointestinal stromal tumors, esophageal early cancer, biliary tract, and liver lesions. The application of AI in EUS diagnosis still has some urgent problems to be solved. First, the development of sensitive AI diagnostic tools requires a large amount of high-quality training data. Second, there is overfitting and bias in the current AI algorithms, leading to poor diagnostic reliability. Third, the value of AI still needs to be determined in prospective studies. Fourth, the ethical risks of AI need to be considered and avoided.

6.
J Clin Med ; 13(11)2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38892819

RESUMEN

Introduction: Pancreatic cancer ranks as the fourth deadliest form of cancer. However, it is essential to note that not all pancreatic masses signal primary malignancy. Therefore, it is imperative to establish the correct differential diagnosis, a process further supported by pre-operative biopsy procedures. This meta-analysis aims to compare the diagnostic performance of two minimally invasive biopsy approaches for pancreatic tissue sampling: percutaneous biopsies guided by computed tomography or ultrasound, and transduodenal biopsies guided by endoscopic ultrasound (EUS). Methods: A systematic literature search was conducted in the MEDLINE and Scopus databases. The included studies analyzed the diagnostic performance of the two biopsy methods, and they were assessed for risk of bias using the Quality Assessment of Diagnostic Accuracy Studies-2 tool. Statistical analysis was carried out using the RevMan and MetaDisc software packages. Results: The statistical analysis of the results demonstrated the superiority of the percutaneous approach. Specifically, the pooled sensitivity, specificity, LR+, LR-and DOR for the percutaneous approach were 0.896 [95% CI: 0.878-0.913], 0.949 [95% CI: 0.892-0.981], 9.70 [95% CI: 5.20-18.09], 0.20 [95% CI: 0.12-0.32] and 68.55 [95% CI: 32.63-143.98], respectively. The corresponding values for EUS-guided biopsies were 0.806 [95% CI: 0.775-0.834], 0.955 [95% CI: 0.926-0.974], 12.04 [95% CI: 2.67-54.17], 0.24 [95% CI: 0.15-0.39] and 52.56 [95% CI: 13.81-200.09], respectively. Nevertheless, it appears that this statistical superiority is also linked to the selection bias favoring larger and hence more readily accessible tumors during percutaneous biopsy procedures. Conclusions: Concisely, our meta-analysis indicates the statistical superiority of the percutaneous approach. However, selecting the optimal biopsy method is complex, influenced by factors like patient and tumor characteristics, clinical resources, and other relevant considerations.

7.
Cancers (Basel) ; 16(9)2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38730610

RESUMEN

INTRODUCTION: Endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA) and endoscopic ultrasound-guided fine-needle biopsy (EUS-FNB) are currently recommended for the pathologic diagnosis of pancreatic solid lesions (PSLs). The application of contrast-enhanced endoscopic ultrasound (ECEUS) could aid the endoscopist during an FNA and/or FNB procedure. CEUS is indeed able to better differentiate the pathologic tissue from the surrounding healthy pancreatic parenchyma and to detect necrotic areas and vessels. OBJECTIVES: Our objective was to evaluate if ECEUS could reduce the number of needle passes and side effects and increase the diagnostic efficacy of FNA and/or FNB. METHODS: A comprehensive literature search of clinical studies was performed to explore if ECEUS-FNA or FNB could increase diagnostic accuracy and reduce the number of needle passes and adverse effects compared to standard EUS-FNA or FNB. In accordance with the study protocol, a qualitative and quantitative analysis of the evidence was planned. RESULTS: The proportion of established diagnoses of ECEUS was 90.9% compared to 88.3% of EUS, with no statistically significant difference (p = 0.14). The diagnosis was made through a single step in 70.9% of ECEUS patients and in 65.3% of EUS patients, without statistical significance (p = 0.24). The incidence of adverse reactions was substantially comparable across both groups (p = 0.89). CONCLUSION: ECEUS-FNA and FNB do not appear superior to standard EUS-FNA and FNB for the diagnosis of pancreatic lesions.

8.
Int J Mol Sci ; 25(6)2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38542201

RESUMEN

Undetermined pancreatic cystic lesion (PCL) differentiation benefits from endoscopic ultrasound (EUS) based on morphology and cyst fluid analysis, but room for new biomarkers exists. Our aim was to assess the intracystic and serum diagnostic value of neutrophil gelatinase-associated lipocalin (Ngal) and interleukin 1 beta (IL-1ß) for differentiation of PCLs. This prospective study included patients from one tertiary hospital, evaluated between April 2018 and May 2020. EUS fine-needle aspiration or pancreatic pseudocysts drainage was the source of PCL intracystic liquid. The final diagnosis was based on surgery or EUS results (morphology, cytology, glucose, and CEA-carcinoembryogenic antigen). The intracystic samples were tested for Ngal, IL-1ß, glucose, and CEA, and serum for Ngal and IL-1ß. We evaluated 63 cysts, 33 pseudocysts, and 30 non-inflammatory cysts. The diagnostic sensitivity and specificity for mucinous PCL was 70.8% and 92.3% for intracystic Ngal (cut-off: 500-800 ng/dL), without correlation with serum Ngal, no matter the inclusion of infected pseudocysts. After exclusion of infected pseudocysts, the sensitivity and specificity for glucose were 87% and 75%, respectively, and for CEA, they were 87.1%, and 96.8%, respectively. Intracystic Ngal shows promise in differentiating mucinous PCLs, but researchers need to conduct further studies to confirm its effectiveness. Intracystic IL-1ß and serum Ngal made no diagnostic contribution.


Asunto(s)
Quiste Pancreático , Neoplasias Pancreáticas , Humanos , Antígeno Carcinoembrionario , Glucosa , Lipocalina 2/análisis , Quiste Pancreático/diagnóstico , Quiste Pancreático/patología , Neoplasias Pancreáticas/patología , Estudios Prospectivos
9.
Postgrad Med ; 136(1): 78-83, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38315145

RESUMEN

OBJECTIVES: Endoscopic ultrasound-guided tissue acquisition (EUS-TA) has been the most common method used for the preoperative cytopathological diagnosis of solid tumors of the pancreas. There are only a few reported cases about the role of endoscopic ultrasound-guided fine needle biopsy (EUS-FNB) in the pre-operative diagnosis of solid pseudopapillary neoplasms (SPN). This study aimed to evaluate the diagnostic yield of EUS-TA,including endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) andEUS-FNB, in patients with SPN. METHODS: We performed a retrospective analysis of patients with EUS-TA for SPN diagnosis in 2 referral centers. The primary outcome was the diagnostic yield of EUS-TA compared to the surgical specimen. RESULTS: Seventy-four patients with SPN of the pancreas were identified. Eighteen had a EUS-TA (10 EUS-FNB and 8 EUS-FNA). The median age of the patients was 31 years (IQR 21-38), and all patients were women. The most common presenting symptom was abdominal pain. Most of the tumors were in the head of the pancreas (9/18; 50%). The median tumor size by EUS was 4.5 cm (min-max 2-15 cm). The most common appearance on EUS was a solid lesion (n = 8/18, 44.4%). A definitive presurgical cytopathological diagnosis was obtained in 16/18 patients (88.8%) with EUS-TA. The sensitivity and positive predictive value of the EUS-TA were 94% each. One patient in the EUS-FNB group developed mild acute pancreatitis. CONCLUSION: The diagnostic yield of the EUS-TA in SPN is high. In most cases, the diagnosis was obtained with the first procedure. No differences in the diagnostic yield or AEs between EUS-FNA vs. EUS-FNB needles were seen.


Asunto(s)
Neoplasias Pancreáticas , Pancreatitis , Humanos , Femenino , Adulto Joven , Adulto , Masculino , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/patología , Estudios Retrospectivos , Enfermedad Aguda , Páncreas/diagnóstico por imagen , Páncreas/patología
10.
Diagnostics (Basel) ; 14(4)2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38396466

RESUMEN

A small tumor size may impact the diagnostic performance of endoscopic ultrasound-guided tissue acquisition (EUS-TA) for diagnosing solid pancreatic lesions (SPLs). We aimed to compare the diagnostic yield of EUS-guided fine-needle aspiration (FNA) and biopsy (FNB) in SPLs with a diameter ≤ 15 mm. Consecutive patients who underwent EUS-TA for SPLs ≤ 15 mm between January 2015 and December 2022 in a tertiary referral center were retrospectively evaluated. The primary endpoint was diagnostic accuracy. The final diagnosis was based on surgical pathology or disease evolution after a minimum follow-up of 6 months. Inadequate samples were all considered false negatives for the study. Secondary outcomes included sample adequacy, factors impacting accuracy, and safety. We included 368 patients (52.4% male; mean age: 60.2 years) who underwent FNA in 72 cases and FNB in 296. The mean size of SPLs was 11.9 ± 2.6 mm. More than three passes were performed in 5.7% and 61.5% of patients in the FNB and FNA groups, respectively (p < 0.0001). FNB outperformed FNA in terms of diagnostic accuracy (89.8% vs. 79.1%, p = 0.013) and sample adequacy (95.9% vs. 86.1%, p < 0.001). On multivariate analysis, using FNA (OR: 2.10, 95% CI: 1.07-4.48) and a final diagnosis (OR: 3.56, 95% CI: 1.82-6.94) of benign conditions negatively impacted accuracy. Overall, the adverse event rate was 0.8%, including one pancreatitis in the FNA group and one pancreatitis and one bleeding in the FNB group, all mild and conservatively managed. EUS-TA for SPLs ≤ 15 mm has a high diagnostic yield and safety. This study suggests the superiority of FNB over FNA, with better performance even with fewer passes performed.

11.
Gastroenterology ; 166(5): 842-858.e5, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38154529

RESUMEN

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) is characterized by desmoplastic stroma surrounding most tumors. Activated stromal fibroblasts, namely cancer-associated fibroblasts (CAFs), play a major role in PDAC progression. We analyzed whether CAFs influence acinar cells and impact PDAC initiation, that is, acinar-to-ductal metaplasia (ADM). ADM connection with PDAC pathophysiology is indicated, but not yet established. We hypothesized that CAF secretome might play a significant role in ADM in PDAC initiation. METHODS: Mouse and human acinar cell organoids, acinar cells cocultured with CAFs and exposed to CAF-conditioned media, acinar cell explants, and CAF cocultures were examined by means of quantitative reverse transcription polymerase chain reaction, RNA sequencing, immunoblotting, and confocal microscopy. Data from liquid chromatography with tandem mass spectrometry analysis of CAF-conditioned medium and RNA sequencing data of acinar cells post-conditioned medium exposure were integrated using bioinformatics tools to identify the molecular mechanism for CAF-induced ADM. Using confocal microscopy, immunoblotting, and quantitative reverse transcription polymerase chain reaction analysis, we validated the depletion of a key signaling axis in the cell line, acinar explant coculture, and mouse cancer-associated fibroblasts (mCAFs). RESULTS: A close association of acino-ductal markers (Ulex europaeus agglutinin 1, amylase, cytokeratin-19) and mCAFs (α-smooth muscle actin) in LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx1Cre (KPC) and LSL-KrasG12D/+; Pdx1Cre (KC) autochthonous progression tumor tissue was observed. Caerulein treatment-induced mCAFs increased cytokeratin-19 and decreased amylase in wild-type and KC pancreas. Likewise, acinar-mCAF cocultures revealed the induction of ductal transdifferentiation in cell line, acinar-organoid, and explant coculture formats in WT and KC mice pancreas. Proteomic and transcriptomic data integration revealed a novel laminin α5/integrinα4/stat3 axis responsible for CAF-mediated acinar-to-ductal cell transdifferentiation. CONCLUSIONS: Results collectively suggest the first evidence for CAF-influenced acino-ductal phenotypic switchover, thus highlighting the tumor microenvironment role in pancreatic carcinogenesis inception.


Asunto(s)
Células Acinares , Fibroblastos Asociados al Cáncer , Carcinoma Ductal Pancreático , Transdiferenciación Celular , Laminina , Neoplasias Pancreáticas , Animales , Humanos , Ratones , Células Acinares/metabolismo , Células Acinares/patología , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/genética , Línea Celular Tumoral , Técnicas de Cocultivo , Medios de Cultivo Condicionados/metabolismo , Metaplasia/patología , Metaplasia/metabolismo , Organoides/metabolismo , Organoides/patología , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/genética , Transducción de Señal , Microambiente Tumoral
12.
Trials ; 24(1): 805, 2023 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-38093381

RESUMEN

BACKGROUND: Studies have shown that the wet suction technique in endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA) generates better histological diagnostic accuracy and specimen quality than the dry suction technique. However, conclusions of wet suction on the diagnostic accuracy of endoscopic ultrasound-guided fine-needle biopsy (EUS-FNB) are still controversial. Besides, the optimal number of passes for EUS-FNB has not been determined. We aimed to design a large multicenter randomized trial to compare the diagnostic accuracy of dry suction versus wet suction technique in solid pancreatic lesions (SPLs) using 22G Franseen needles and determine the optimal number of passes required for EUS-FNB. METHODS: This is a multi-center open-label, randomized controlled non-inferiority trial with two parallel groups. Two hundred patients with SPLs will undergo EUS-FNB using 22G Franseen needles in 4 tertiary hospitals in China and will be randomly assigned to the dry suction group and wet suction group in a ratio of 1:1. The primary endpoint is diagnostic accuracy. Secondary endpoints include the optimal number of needle passes, sensitivity, specificity, specimen quality, cytological diagnoses, time of the procedure, and incidence of complications. DISCUSSION: This study has been designed to determine (i) whether EUS-FNB using 22G Franseen needle with dry suction is non-inferior to wet suction in terms of diagnostic accuracy and (ii) the optimal number of passes during EUS-FNB of SPLs using 22G Franseen needle. TRIAL REGISTRATION: ClinicalTrials.gov NCT05549856. Registered on September 22, 2022.


Asunto(s)
Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/patología , Succión , Páncreas/patología , Biopsia Guiada por Imagen , Biopsia por Aspiración con Aguja Fina Guiada por Ultrasonido Endoscópico/métodos , Ensayos Clínicos Controlados Aleatorios como Asunto , Estudios Multicéntricos como Asunto
13.
Artículo en Inglés | MEDLINE | ID: mdl-37914883

RESUMEN

This article provides an extensive review of the advancements and future perspectives related to endoscopic ultrasound-guided tissue acquisition (EUS-TA) for the diagnosis of solid pancreatic lesions (SPLs). EUS-TA, including fine-needle aspiration (EUS-FNA) and fine-needle biopsy (EUS-FNB), has revolutionized the collection of specimens from intra-abdominal organs, including the pancreas. Improvements in the design of needles, collection methods, and specimen processing techniques have improved the diagnostic performance. This review highlights the latest findings regarding needle evolution, actuation number, sampling methods, specimen evaluation techniques, application of artificial intelligence (AI) for diagnostic purposes, and use of comprehensive genomic profiling (CGP). It acknowledges the rising use of Franseen and fork-tip needles for EUS-FNB and emphasizes that the optimal number of actuations requires further study. Methods such as the door-knocking and fanning techniques have shown promise for increasing diagnostic performance. Macroscopic on-site evaluation (MOSE) is presented as a practical rapid specimen evaluation method, and the integration of AI is identified as a potentially impactful development. The study also underscores the importance of optimal sampling for CGP, which can enhance the precision of cancer treatment. Ongoing research and technological innovations will further improve the accuracy and efficacy of EUS-TA.

14.
Front Pediatr ; 11: 1278463, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38027294

RESUMEN

Pancreaticopleural fistula (PPF) is a rare but serious complication caused by pancreatic lesions that presents primarily with respiratory tract symptoms and pleural effusion. We report a paediatric case of PPF without any respiratory symptoms throughout the course of the disease, including cough or shortness of breath, with only a bulging chest as the first symptom. Imaging revealed a large left pleural effusion and Magnetic Resonance Cholangiopancreatography (MRCP) revealed a fistula formed between the pancreatic tail and the pleural cavity, which penetrated the diaphragm and opened in the central tendon of the diaphragm. The patient eventually underwent resection of the pancreatic tail lesion and repair of the diaphragmatic fistula and recovered soon thereafter.

15.
Diagnostics (Basel) ; 13(19)2023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-37835797

RESUMEN

Solid pancreatic lesions (SPLs) encompass a variety of benign and malignant diseases and accurate diagnosis is crucial for guiding appropriate treatment decisions. Endoscopic ultrasonography-guided fine-needle aspiration/biopsy (EUS-FNA/B) serves as a front-line diagnostic tool for pancreatic mass lesions and is widely used in clinical practice. Artificial intelligence (AI) is a mathematical technique that automates the learning and recognition of data patterns. Its strong self-learning ability and unbiased nature have led to its gradual adoption in the medical field. In this paper, we describe the fundamentals of AI and provide a summary of reports on AI in EUS-FNA/B to help endoscopists understand and realize its potential in improving pathological diagnosis and guiding targeted EUS-FNA/B. However, AI models have limitations and shortages that need to be addressed before clinical use. Furthermore, as most AI studies are retrospective, large-scale prospective clinical trials are necessary to evaluate their clinical usefulness accurately. Although AI in EUS-FNA/B is still in its infancy, the constant input of clinical data and the advancements in computer technology are expected to make computer-aided diagnosis and treatment more feasible.

16.
Artículo en Inglés | MEDLINE | ID: mdl-37554973

RESUMEN

Background: Despite advancement in imaging techniques, the diagnosis of solid pancreatic lesions (SPLs) remains challenging. The latest advancement in elastography permits the quantitative measurements of the average elasticity of a lesion. Therefore, our main aim of this study was to determine the utility of endoscopic ultrasound-guided elastography (EUS-EG) and strain ratio (EUS-SR) in predicting SPLs. Materials and methods: This cross-sectional study was performed at the Department of Hepatogastroenterology, Sindh Institute of Urology and Transplantation. All patients with radiological diagnosis of SPLs underwent EUS-EG, followed by strain ratio (SR) measurement and targeted pancreatic fine needle lesion biopsy (FNB). Area under the receiver operating curve (AUROC) was obtained for SR and combined elastography and SR and at an optimal cutoff, diagnostic accuracy was obtained in predicting the nature of SPLs. Results: A total of 52 patients were included in this study. Out of them, 32 (61.5%) patients were males while 20 (38.5%) were females. The mean age was 50.8 ± 12.5 years. Twenty-four (46.2%) patients had malignant pancreatic lesions. Among malignant lesions, the most common etiology was pancreatic adenocarcinoma seen in 18 (34.6%) patients. Out of 28 (53.8%) patients with benign lesions, 14 (26.9%) patients had inflammatory disease. Area under the receiver operating curve was obtained for both SR alone and SR combined with elastography score in differentiating benign from malignant SPLs which was 0.832 (p-value < 0.001) for SR alone and a slightly higher for combined SR with elastography (AUROC-0.839)(p-value < 0.001). At an optimal cutoff of SR of >17, the sensitivity was 94.8% and the diagnostic accuracy was 74% in predicting SPLs. While, when SR and elastography were combined together, the sensitivity increased to 96% with a diagnostic accuracy of 75%. Conclusion: Combined EUS-EG and SR were accurate in diagnosing malignant pancreatic lesions with a diagnostic accuracy of 75% providing additional diagnostics information before biopsy. However, multicentric studies with larger sample sizes are required for the validation of our results to determine the utility and diagnostic accuracy of EUS-SR in defining the characteristic of pancreatic lesions. How to cite this article: Bajaj K, Yaseen T, Tasneem AA, et al. Role of Endoscopic Ultrasound in Predicting Solid Pancreatic Lesions Using Strain Ratio and Elastography. Euroasian J Hepato-Gastroenterol 2023;13(1):1-4.

17.
Radiol Med ; 128(9): 1035-1046, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37515631

RESUMEN

PURPOSE: The pancreatic cancer (PC) is the 4th leading cancer-related death, becoming the second one by 2030, with a 5 year survival rate of 8%. Considering its increased incidence in high-risk categories compared to the general population, we aimed to validate a non-contrast MR protocol, to detect PC in its earliest phase, which could be suitable as a screening tool in high-risk patients. MATERIALS AND METHODS: In this retrospective study, we selected 200 patients (> 40 years) from our radiological database, which performed upper abdominal MRI between 2012 and 2017. 100 were negative for pancreatic lesions and 100 positive for pancreatic lesion (< 30 mm). The latter group included: 40 PDAC (pancreatic adenocarcinoma), 42 BD-IPMN (Branch Duct- Intraductal Papillary Mucinous Neoplasm), 10 PNET(pancreatic neuroendocrine tumor), 4 SCN(serous cystic neoplasm), 3 IPS(intrapancreatic spleen), 1 MCN(mucinous cystic neoplasm). Three readers (R1, R2 and R3) with a high, medium and low experience, respectively, analysed, first, the non-contrast MR sequences (single-shot T2w breath-hold, GE T1w FS, DWI and 2D/3D MRCP), and then the standard MR protocol, independently, randomly and anonymously. Readers identified or excluded the presence of pancreatic lesion, in both reading sessions. These results were compared with the histopathological diagnosis, and then divided into 3 different classes of lesions: all lesions, pancreatic adenocarcinoma and solid lesion. Mcnemar's test was used to compare the results. The inter-observer agreement was determined according to the kappa statistic in both protocols, and then the inter-protocol agreement was calculated. RESULTS: The non-contrast MR protocol has reached statistical parameters values ranging between 83% in SE (sensitivity) by R3 and 99% in NPV (negative predictive value) by R1. The standard MR protocol has reported slight increasing statistical parameters compared to those of the proposed one. However, there are not significant statistical differences between the both protocols. The proposed non-contrast MR protocol has reported the highest NPVs in the PDAC group detection (R1: 99%, R2: 99%, R3: 98%). In all groups of lesions, the agreement between the two protocols was excellent for each Reader ranging from 96 to 98%. CONCLUSION: The proposed non-contrast MR protocol showed high PC detection values and a time execution ≤ 20 min. Therefore, it can be proposed as a screening tool in high-risk patients.


Asunto(s)
Adenocarcinoma , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/diagnóstico por imagen , Adenocarcinoma/diagnóstico por imagen , Estudios Retrospectivos , Imagen por Resonancia Magnética/métodos , Carcinoma Ductal Pancreático/diagnóstico por imagen , Carcinoma Ductal Pancreático/patología , Neoplasias Pancreáticas
18.
United European Gastroenterol J ; 11(7): 601-611, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37435855

RESUMEN

BACKGROUND: Surveillance of pancreatic cysts focuses on the detection of (mostly morphologic) features warranting surgery. European guidelines consider elevated CA19.9 as a relative indication for surgery. We aimed to evaluate the role of CA19.9 monitoring for early detection and management in a cyst surveillance population. METHODS: The PACYFIC-registry is a prospective collaboration that investigates the yield of pancreatic cyst surveillance performed at the discretion of the treating physician. We included participants for whom at least one serum CA19.9 value was determined by a minimum follow-up of 12 months. RESULTS: Of 1865 PACYFIC participants, 685 met the inclusion criteria for this study (mean age 67 years, SD 10; 61% female). During a median follow-up of 25 months (IQR 24, 1966 visits), 29 participants developed high-grade dysplasia (HGD) or pancreatic cancer. At baseline, CA19.9 ranged from 1 to 591 kU/L (median 10 kU/L [IQR 14]), and was elevated (≥37 kU/L) in 64 participants (9%). During 191 of 1966 visits (10%), an elevated CA19.9 was detected, and these visits more often led to an intensified follow-up (42%) than those without an elevated CA19.9 (27%; p < 0.001). An elevated CA19.9 was the sole reason for surgery in five participants with benign disease (10%). The baseline CA19.9 value was (as continuous or dichotomous variable at the 37 kU/L threshold) not independently associated with HGD or pancreatic cancer development, whilst a CA19.9 of ≥ 133 kU/L was (HR 3.8, 95% CI 1.1-13, p = 0.03). CONCLUSIONS: In this pancreatic cyst surveillance cohort, CA19.9 monitoring caused substantial harm by shortening surveillance intervals (and performance of unnecessary surgery). The current CA19.9 cutoff was not predictive of HGD and pancreatic cancer, whereas a higher cutoff may decrease false-positive values. The role of CA19.9 monitoring should be critically appraised prior to implementation in surveillance programs and guidelines.


Asunto(s)
Quiste Pancreático , Neoplasias Pancreáticas , Humanos , Femenino , Anciano , Masculino , Estudios Prospectivos , Antígeno CA-19-9 , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/epidemiología , Quiste Pancreático/diagnóstico , Quiste Pancreático/cirugía , Neoplasias Pancreáticas
19.
Artículo en Inglés | MEDLINE | ID: mdl-37341913

RESUMEN

BACKGROUND AND AIM: Endoscopic ultrasound-guided through-the-needle biopsy (EUS-TTNB) has been used over the past few years to increase diagnostic accuracy for pancreatic cystic lesions (PCLs). However, many concerns remain regarding its widespread use. This systematic review and meta-analysis aimed to pool the data from high-quality studies to evaluate the utility of EUS-TTNB in diagnosing PCLs. METHODS: Electronic databases (PubMed, Embase, and Cochrane Library) from January 2010 through October 2022 were searched for publications addressing the diagnostic performance of EUS-TTNB in the diagnosis of pancreatic cystic lesions. Pooled proportions were calculated using fixed (inverse variance) and random-effects (DerSimonian-Laird) models. RESULTS: The initial search identified 635 studies, of which 35 relevant articles were reviewed. We extracted data from 11 studies that met the inclusion criterion, comprising a total of 575 patients. Mean patient age was 62.25 years ± 6.12 with females constituting 61.39% of the study population. Pooled sensitivity of EUS-TTNB in differentiating a PCL as neoplastic or non-neoplastic was 76.60% (95% CI = 72.60-80. 30). For the same indication, EUS TTNB had a pooled specificity of 98.90% (95% CI = 93.80-100.00). The positive likelihood ratio was 10.28 (95% CI = 4.77-22.15), and the negative likelihood ratio was 0.26 (95% CI = 0.22-0.31). The pooled diagnostic odds ratio for EUS-TTNB in diagnosing PCLs as malignant/pre-malignant vs. non-malignant was 41.34 (95% CI = 17.42-98.08). Pooled adverse event rates were 3.04% (95% CI = 1.83-4.54) for pancreatitis, 4.02% (95% CI = 2.61-5.72) for intra-cystic bleeding, 0.94% (95% CI = 0.33-1.86) for fever, and 1.73% (95% CI = 0.85-2.91) for other minor events. CONCLUSIONS: EUS-TTNB has good sensitivity with excellent specificity in accurately classifying PCLs as neoplastic or non-neoplastic. Adding EUS-TTNB to EUS-FNA increases the accuracy of EUS-guided approach in diagnosing PCLs. However, it could significantly increase the risk of post-procedural pancreatitis.

20.
Acta Cytol ; 67(3): 240-247, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37263259

RESUMEN

INTRODUCTION: Early detection and accurate pathological assessment are critical to improving prognosis of pancreatic cancer. EUS has been widely used in diagnosing pancreatic lesions and can obtain histological diagnosis by endoscopic ultrasound-guided fine needle aspiration (EUS-FNA). However, comprehensive assessment of the interobserver agreement (IOA) among cytopathologists evaluating EUS-FNA specimens is still limited. Therefore, this study evaluated IOA among cytopathologists for EUS-FNA specimens of solid pancreatic lesions, especially in false-negative cases of cytological diagnosis and analyzed the factors that influence cytological diagnosis of EUS-FNA so as to improve the diagnostic efficiency of EUS-FNA. METHODS: We retrieved EUS-FNA samples of pancreatic solid lesions from 2017 to 2021 and collected their clinical/cytological data. Two cytopathologists independently reviewed these cases using a quoted, novel standardized cytology scoring tool. Ultimately, we calculated IOA among cytopathologists and performed a binary logistic regression analysis to evaluate factors influencing the cytological diagnosis of EUS-FNA. RESULTS: 161 patients were included, and 60 cases with a clinical diagnosis of pancreatic cancer but a cytological diagnosis of benign and atypical constituted the false-negative group. IOAs for cytological diagnosis of overall patients and the false-negative group were in perfect/moderate agreement with Kendall's W values of 0.896 and 0.462, respectively. The number of diagnostic cells in the scoring tool had the highest level of agreement (κ = 0.721) for overall patients. There was at best moderate agreement on other quantity and quality parameters for both all cases and false-negative group. Logistic regression analysis showed the number of diagnostic cells (OR = 6.110, p < 0.05) and amount of blood (OR = 0.320, p < 0.05) could influence cytological diagnosis. CONCLUSIONS: The false-negative rate of our study as high as 37.26% (60/161) is mainly related to strict standards of cytopathologists, and their ability to standardize pancreatic cytology is still improving. Suboptimal agreement among cytopathologists for cytological diagnosis and the number of diagnostic cells may be associated with the occurrence of false-negative diagnosis. Further regression analysis confirmed that the number of diagnostic cells and obscuring blood were important factors in cytological diagnosis. Therefore, refinement of cytological diagnostic criteria, standardization of specimen quality evaluation, and training of cytopathologists may improve the agreement of cytopathologists, thus improving the repeatability of cytological diagnosis and reducing the occurrence of false-negative events.


Asunto(s)
Biopsia por Aspiración con Aguja Fina Guiada por Ultrasonido Endoscópico , Neoplasias Pancreáticas , Humanos , Variaciones Dependientes del Observador , Páncreas/patología , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA