1
|
Leupold M, Chen W, Esnakula AK, Frankel WL, Culp S, Hart PA, Abdelbaki A, Shah ZK, Park E, Lee P, Ramsey ML, Han S, Shah H, Burlen J, Papachristou GI, Cruz-Monserrate Z, Dillhoff M, Cloyd JM, Pawlik TM, Krishna SG. Interobserver agreement in dysplasia grading of intraductal papillary mucinous neoplasms: performance of Kyoto guidelines and optimization of endomicroscopy biomarkers through pathology reclassification. Gastrointest Endosc 2025; 101:1155-1165.e6. [PMID: 39557199 DOI: 10.1016/j.gie.2024.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND AND AIMS Interobserver agreement (IOA) among pancreaticobiliary (PB) pathologists in evaluating high-grade dysplasia and/or invasive carcinoma (HGD-IC) of intraductal papillary mucinous neoplasms (IPMNs) remains understudied. EUS-guided needle-based confocal endomicroscopy (nCLE) can evaluate papillary architecture in branch duct IPMNs. We assessed IOA among PB pathologists in classifying dysplasia in resected IPMNs and compared the performance of the Kyoto guidelines' high-risk stigmata (HRS) and presurgical EUS-nCLE against reclassified pathology. METHODS Participants in prospective clinical trials (2015-2023) with resected IPMNs were included. Blinded PB pathologists independently reviewed histopathology, achieving a consensus diagnosis. The accuracies of cyst fluid next-generation sequencing analysis, EUS-nCLE, and Kyoto HRS in predicting HGD-IC were compared with the reclassified pathology. RESULTS Among 64 participants, 25 (39%) exhibited HGD-IC (17 HGD, 8 invasive carcinoma). Disagreements occurred in 14% of cases with substantial IOA (κ = 0.70; 95% confidence interval, 0.53-0.88) between 2 PB pathologists for differentiating HGD-IC versus low-grade dysplasia (LGD). To detect HGD-IC, the sensitivity, specificity, and accuracy of Kyoto HRS and EUS-nCLE were 52%, 95%, 78% and 68%, 87%, 80%, respectively. Integrating nCLE with Kyoto HRS improved sensitivity to 80%, with specificity and accuracy at 82% and 81%, respectively. The sensitivity, specificity, and accuracy of next-generation sequencing (n = 47) to detect HGD-IC were 6.3%, 100%, and 68%, respectively. A unique subset of IPMNs were identified in all (n = 8, P = .01) cases where presurgical EUS-nCLE underestimated dysplasia revealing a distinct micropapillary architecture on postsurgical histopathology. CONCLUSIONS Despite substantial IOA among experienced PB pathologists, a second pathologist's review may be warranted for dysplasia classification in IPMNs under certain circumstances. Incorporating an imaging biomarker such as EUS-nCLE with Kyoto HRS improves sensitivity for HGD-IC without sacrificing accuracy.
Collapse
Affiliation(s)
- Matthew Leupold
- Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Wei Chen
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Ashwini K Esnakula
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Wendy L Frankel
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Stacey Culp
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Philip A Hart
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Ahmed Abdelbaki
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Zarine K Shah
- Department of Radiology, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Erica Park
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Peter Lee
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Mitchell L Ramsey
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Samuel Han
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Hamza Shah
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Jordan Burlen
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Georgios I Papachristou
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Mary Dillhoff
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Jordan M Cloyd
- The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Timothy M Pawlik
- The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Somashekar G Krishna
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
2
|
Iyer MK, Fletcher A, Okoye JO, Shi C, Chen F, Kanu E, Eckhoff AM, Bao M, di Magliano MP, Frankel TL, Chinnaiyan AM, Nussbaum DP, Allen PJ. Spatial Transcriptomics of Intraductal Papillary Mucinous Neoplasms Reveals Divergent Indolent and Malignant States. Clin Cancer Res 2025; 31:1796-1808. [PMID: 39969959 PMCID: PMC12045729 DOI: 10.1158/1078-0432.ccr-24-1529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/20/2024] [Accepted: 02/17/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE Intraductal papillary mucinous neoplasms (IPMN) occur in 5% to 10% of the population, but only a small minority progress to pancreatic ductal adenocarcinoma (PDAC). The lack of accurate predictors of high-risk disease leads to both unnecessary operations for indolent neoplasms and missed diagnoses of PDAC. Digital spatial RNA profiling (DSP-RNA) provides an opportunity to define and associate transcriptomic states with cancer risk. EXPERIMENTAL DESIGN We performed whole-transcriptome DSP-RNA profiling on 10 IPMN specimens encompassing the spectrum of dysplastic changes from normal duct to cancer. Epithelial regions within each tissue were annotated as normal duct, low-grade dysplasia, high-grade dysplasia, or invasive carcinoma. The resulting digital gene expression data were analyzed with R/Bioconductor. RESULTS Our analysis uncovered three distinct epithelial transcriptomic states-"normal-like" (cNL), "low risk" (cLR), and "high risk" (cHR)-which were significantly associated with pathologic grade. Furthermore, the three states were significantly correlated with the exocrine, classical, and basal-like molecular subtypes described in PDAC. Specifically, exocrine function diminished in cHR, classical activation distinguished neoplasia (cLR and cHR) from cNL, and basal-like genes were specifically upregulated in cHR. Intriguingly, markers of cHR were detected in normal duct and low-grade dysplasia regions from specimens with PDAC but not from specimens containing only low-grade IPMN. CONCLUSIONS DSP-RNA of IPMN revealed low-risk (indolent) and high-risk (malignant) expression programs that correlated with the activity of exocrine and basal-like PDAC signatures, respectively, and distinguished pathologically low-grade specimens from malignant specimens. These findings contextualize IPMN pathogenesis and have the potential to improve risk stratification.
Collapse
Affiliation(s)
- Matthew K. Iyer
- Department of Surgery, Duke University; Durham, North Carolina
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Ashley Fletcher
- Department of Surgery, Duke University; Durham, North Carolina
| | - Jude Ogechukwu Okoye
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Department of Histopathology, Nnamdi Azikiwe University, Nnewi, Nigeria
| | - Chanjuan Shi
- Department of Pathology, Duke University; Durham, North Carolina
| | - Fengming Chen
- Department of Pathology, Duke University; Durham, North Carolina
| | - Elishama Kanu
- Department of Surgery, Duke University; Durham, North Carolina
| | | | - Matthew Bao
- Department of Surgery, Duke University; Durham, North Carolina
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | | | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, Duke University; Durham, North Carolina
- Department of Urology, University of Michigan, Ann Arbor, Michigan
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| | | | - Peter J. Allen
- Department of Surgery, Duke University; Durham, North Carolina
| |
Collapse
|
3
|
Hamada T, Oyama H, Takahara N, Nakai Y, Fujishiro M. Role of Endoscopy in Clinical Management of Intraductal Papillary Mucinous Neoplasms. J Gastroenterol Hepatol 2025; 40:1045-1058. [PMID: 40159838 PMCID: PMC12062927 DOI: 10.1111/jgh.16938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/21/2025] [Accepted: 03/09/2025] [Indexed: 04/02/2025]
Abstract
Intraductal papillary mucinous neoplasm (IPMN) of the pancreas is a well-recognized precursor of pancreatic carcinoma. Along with cross-sectional abdominal imaging tests, endoscopic examinations remain the cornerstone in the diagnosis of pancreatic cysts, early detection of IPMN-derived carcinomas, and risk stratification of patients with IPMNs for subsequent surveillance strategies. In particular, endoscopic ultrasound (EUS) facilitates the optimal patient management by providing high-resolution morphological information, and the contrast-enhanced harmonic mode may further enhance diagnostic accuracy. EUS-guided fine-needle aspiration for solid mass and/or cyst fluid is considered for pathological and molecular examinations for the diagnosis of pancreatic cysts and malignancy. Emerging evidence suggests the usefulness of through-the-needle biopsy and confocal laser microendoscopy in this setting. In addition to the undoubtful diagnostic utility, recent studies have demonstrated the potential effect of endoscopic interventions (i.e., ablation) on the control of IPMNs. Despite the increasing role of endoscopy in the clinical management of IPMNs, there remains a gap in our understanding of how to utilize endoscopy in the personalized care for patients with IPMNs (e.g., the optimal interval of EUS) and the prevention of deaths due to pancreatic carcinomas developing concomitantly with IPMNs. This review summarizes the current evidence on the role of endoscopy in both the diagnostic and therapeutic landscapes of clinical management of IPMNs and identifies key clinical unmet needs that should be addressed in future research. Combined with emerging technologies (e.g., artificial intelligence and high-throughput molecular profiling), endoscopy would offer more effective and tailored management strategies for patients with IPMNs.
Collapse
Affiliation(s)
- Tsuyoshi Hamada
- Graduate School of MedicineDepartment of GastroenterologyThe University of TokyoTokyoJapan
- The Cancer Institute HospitalDepartment of Hepato‐Biliary‐Pancreatic MedicineJapanese Foundation for Cancer ResearchTokyoJapan
| | - Hiroki Oyama
- Graduate School of MedicineDepartment of GastroenterologyThe University of TokyoTokyoJapan
| | - Naminatsu Takahara
- Graduate School of MedicineDepartment of GastroenterologyThe University of TokyoTokyoJapan
| | - Yousuke Nakai
- Graduate School of MedicineDepartment of GastroenterologyThe University of TokyoTokyoJapan
- Institute of GastroenterologyDepartment of Internal MedicineTokyo Women's Medical UniversityTokyoJapan
| | - Mitsuhiro Fujishiro
- Graduate School of MedicineDepartment of GastroenterologyThe University of TokyoTokyoJapan
| |
Collapse
|
4
|
Mishra A, Hunold TM, Peddu DK, Philips GM, Wamsteker EJ, Kwon RS, Schulman AR, Shi J, Carpenter ES, Machicado JD. Histologic Diagnosis of Pancreatic Cystic Lesions with Endoscopic Ultrasound Fine Needle Biopsy and Impact on Management Decisions. Dig Dis Sci 2025:10.1007/s10620-025-09056-1. [PMID: 40261565 DOI: 10.1007/s10620-025-09056-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE Endoscopic ultrasound with fine needle biopsy (EUS-FNB) has not been well studied in pancreatic cystic lesions (PCLs). This study evaluates the diagnostic performance of EUS-FNB for PCLs and its impact on management decisions. METHODS We conducted a single-center, retrospective study of patients who had EUS-FNB between March 2016 and February 2024. We included patients with ≥ 6-month follow-up and excluded those with a solid pancreatic mass. We obtained clinical, radiologic, endoscopic, surgical, laboratory, and pathology data from chart review. We evaluated: (A) diagnostic yield; (B) predictors of diagnostic FNB; (C) diagnostic accuracy compared to surgical histopathology; (D) appropriateness of management decisions; and (E) adverse events. We compared the appropriateness of management decisions between diagnostic and non-diagnostic FNB. RESULTS 100 subjects underwent EUS-FNB for PCLs (56% microcystic or with mural nodule). FNB yielded a histologic diagnosis in 60% of sampled lesions. Performing 2 or more needle passes was the only significant predictor of a diagnostic FNB (p = 0.02). Compared to surgical histopathology (n = 21), FNB needles highly accurately diagnosed specific cyst types (IPMN = 85.7%, MCN = 90.5%, SCA = 95.2%, NET = 95.2%, SPN = 100%) and malignant PCLs (accuracy = 81.0%; specificity = 100%; sensitivity = 72.7%). There was a 7.2-fold increase of appropriate management decisions when FNB was diagnostic vs. non-diagnostic (p < 0.001). This was due to improvement in surveillance discontinuation for benign cysts and in appropriate surgical resection for malignant PCLs (p < 0.001). Post-FNB pancreatitis occurred in 4% of patients. CONCLUSION EUS-FNB accurately diagnoses different PCL types and their degree of neoplasia, leading to more appropriate management decisions. Future prospective studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Ankit Mishra
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Thomas M Hunold
- Division of Gastroenterology and Hepatology, University of Michigan, 1500 E Medical Center Dr, Floor 3 Reception D, Ann Arbor, MI, 48109, USA
| | - Dhiraj K Peddu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - George M Philips
- Division of Gastroenterology and Hepatology, University of Michigan, 1500 E Medical Center Dr, Floor 3 Reception D, Ann Arbor, MI, 48109, USA
| | - Erik-Jan Wamsteker
- Division of Gastroenterology and Hepatology, University of Michigan, 1500 E Medical Center Dr, Floor 3 Reception D, Ann Arbor, MI, 48109, USA
| | - Richard S Kwon
- Division of Gastroenterology and Hepatology, University of Michigan, 1500 E Medical Center Dr, Floor 3 Reception D, Ann Arbor, MI, 48109, USA
| | - Allison R Schulman
- Division of Gastroenterology and Hepatology, University of Michigan, 1500 E Medical Center Dr, Floor 3 Reception D, Ann Arbor, MI, 48109, USA
| | - Jiaqi Shi
- Department of Pathology and Clinical Labs, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Eileen S Carpenter
- Division of Gastroenterology and Hepatology, University of Michigan, 1500 E Medical Center Dr, Floor 3 Reception D, Ann Arbor, MI, 48109, USA
| | - Jorge D Machicado
- Division of Gastroenterology and Hepatology, University of Michigan, 1500 E Medical Center Dr, Floor 3 Reception D, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
5
|
Facciorusso A, Arvanitakis M, Crinò SF, Fabbri C, Fornelli A, Leeds J, Archibugi L, Carrara S, Dhar J, Gkolfakis P, Haugk B, Iglesias Garcia J, Napoleon B, Papanikolaou IS, Seicean A, Stassen PMC, Vilmann P, Tham TC, Fuccio L. Endoscopic ultrasound-guided tissue sampling: European Society of Gastrointestinal Endoscopy (ESGE) Technical and Technology Review. Endoscopy 2025; 57:390-418. [PMID: 40015316 DOI: 10.1055/a-2524-2596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
This Technical and Technology Review from the European Society of Gastrointestinal Endoscopy (ESGE) represents an update of the previous document on the technical aspects of endoscopic ultrasound (EUS)-guided sampling in gastroenterology, including the available types of needle, technical aspects of tissue sampling, new devices, and specimen handling and processing. Among the most important new recommendations are:ESGE recommends end-cutting fine-needle biopsy (FNB) needles over reverse-bevel FNB or fine-needle aspiration (FNA) needles for tissue sampling of solid pancreatic lesions; FNA may still have a role when rapid on-site evaluation (ROSE) is available.ESGE recommends EUS-FNB or mucosal incision-assisted biopsy (MIAB) equally for tissue sampling of subepithelial lesions ≥20 mm in size. MIAB could represent the first choice for smaller lesions (<20 mm) if proper expertise is available.ESGE does not recommend the use of antibiotic prophylaxis before EUS-guided tissue sampling of solid masses and EUS-FNA of pancreatic cystic lesions.
Collapse
Affiliation(s)
- Antonio Facciorusso
- Department of Experimental Medicine, Section of Gastroenterology, University of Salento, Lecce, Italy
| | - Marianna Arvanitakis
- Department of Gastroenterology, Digestive Oncology and Hepatopancreatology, HUB Hôpital Erasme, Brussels, Belgium
| | - Stefano Francesco Crinò
- Department of Medicine, Gastroenterology and Digestive Endoscopy Unit, The Pancreas Institute, University Hospital of Verona, Verona, Italy
| | - Carlo Fabbri
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, AUSL Romagna, Forlì-Cesena, Italy
| | - Adele Fornelli
- Pathology Unit, Ospedale Maggiore "C.A. Pizzardi", AUSL Bologna, Bologna, Italy
| | - John Leeds
- Department of Gastroenterology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Livia Archibugi
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Carrara
- Department of Biomedical Sciences, Humanitas Pieve Emanuele University, Milan, Italy
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Jahnvi Dhar
- Department of Gastroenterology and Hepatology, Punjab Institute of Liver and Biliary Sciences, Mohali, India
| | - Paraskevas Gkolfakis
- Department of Gastroenterology, "Konstantopoulio-Patision" General Hospital of Nea Ionia, Athens, Greece
| | - Beate Haugk
- Department of Cellular Pathology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Julio Iglesias Garcia
- Department of Gastroenterology and Hepatology, Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela, Santiago, Spain
| | - Bertrand Napoleon
- Department of Gastroenterology, Hôpital privé Jean Mermoz, Lyon, France
| | - Ioannis S Papanikolaou
- Hepatogastroenterology Unit, Second Department of Propaedeutic Internal Medicine, Medical School, National and Kapodastrian University of Athens, Attikon University General Hospital, Athens, Greece
| | - Andrada Seicean
- Department of Gastroenterology, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Pauline M C Stassen
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Peter Vilmann
- Gastroenterology Unit, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Tony C Tham
- Division of Gastroenterology, Ulster Hospital, Belfast, Northern Ireland
| | - Lorenzo Fuccio
- Department of Medical Sciences and Surgery, University of Bologna, Bologna, Italy
| |
Collapse
|
6
|
Ohno E, Kuzuya T, Kawabe N, Nakaoka K, Tanaka H, Nakano T, Funasaka K, Miyahara R, Hashimoto S, Hirooka Y. Current status of endoscopic ultrasound in the diagnosis of intraductal papillary mucinous neoplasms. DEN OPEN 2025; 5:e413. [PMID: 39040523 PMCID: PMC11260769 DOI: 10.1002/deo2.413] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/02/2024] [Accepted: 07/06/2024] [Indexed: 07/24/2024]
Abstract
The new Kyoto guidelines for the management of intraductal papillary mucinous neoplasm (IPMN) provide evidence-based recommendations for the diagnosis and treatment of IPMN. Endoscopic ultrasonography (EUS) is a diagnostic modality with a high spatial resolution that allows detailed observation and obtaining cyst fluid or tissue samples via EUS-guided fine needle aspiration (EUS-FNA). Currently, EUS is an indispensable examination method for the diagnosis of pancreatic diseases. On the other hand, there have been concerns that EUS imaging tends to be highly operator-dependent, and may lack objectivity. Previous guidelines have assigned EUS as an option for patients with worrisome features. However, recent reports indicate that the sensitivity of EUS for the diagnosis of mural nodules (MNs) is more than 90%, comparable or superior to that of contrast-enhanced computed tomography or magnetic resonance cholangiopancreatography. The specific advantages of EUS in the diagnosis of IPMN are: (1) high spatial resolution imaging for the diagnosis of MNs, (2) contrast-enhanced EUS for differentiation of intra-cystic MNs from mucous clots, and (3) pathological diagnosis using EUS-FNA and differential diagnosis of a pancreatic cystic tumor by cystic fluid analysis. In order to utilize EUS in the diagnosis of IPMN, endoscopists are required to have the skills to provide sufficiently objective imaging findings.
Collapse
Affiliation(s)
- Eizaburo Ohno
- Department of Gastroenterology and HepatologyFujita Health University School of MedicineAichiJapan
| | - Teiji Kuzuya
- Department of Gastroenterology and HepatologyFujita Health University School of MedicineAichiJapan
| | - Naoto Kawabe
- Department of Gastroenterology and HepatologyFujita Health University School of MedicineAichiJapan
| | - Kazunori Nakaoka
- Department of Gastroenterology and HepatologyFujita Health University School of MedicineAichiJapan
| | - Hiroyuki Tanaka
- Department of Gastroenterology and HepatologyFujita Health University School of MedicineAichiJapan
| | - Takuji Nakano
- Department of Gastroenterology and HepatologyFujita Health University School of MedicineAichiJapan
| | - Kohei Funasaka
- Department of Gastroenterology and HepatologyFujita Health University School of MedicineAichiJapan
| | - Ryoji Miyahara
- Department of Gastroenterology and HepatologyFujita Health University School of MedicineAichiJapan
| | - Senju Hashimoto
- Department of Gastroenterology and HepatologyFujita Health University Bantane HospitalAichiJapan
| | - Yoshiki Hirooka
- Department of Gastroenterology and HepatologyFujita Health University School of MedicineAichiJapan
| |
Collapse
|
7
|
Maher MH, Treekitkarnmongkol W, Ghatak S, Dai J, Liu S, Nguyen T, Duose DY, Kim MP, Hu TY, Hurd MW, Paris PL, Kirkwood KS, Maitra A, Luthra R, Sen S, Roy-Chowdhuri S. An integrated multi-omics biomarker approach using molecular profiling and microRNAs for evaluation of pancreatic cyst fluid. Cancer Cytopathol 2025; 133:e70008. [PMID: 40106268 DOI: 10.1002/cncy.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/07/2025] [Accepted: 01/17/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Classification and risk stratification of pancreatic cysts are challenging because of limited radiographic and cytomorphologic features. Although molecular profiling has emerged as an ancillary test for pancreatic cyst fluid (PCF), additional high-sensitivity and -specificity biomarkers are still needed for improved classification. METHODS In this study, PCF from 93 patients, including intraductal papillary mucinous neoplasms (n = 65), mucinous cystic neoplasms (n = 9), serous cystadenomas (n = 9), pancreatic cyst not otherwise specified (n = 8), and pseudocysts (n = 2), were evaluated for biomarkers. Molecular profiling by next-generation sequencing was performed, and a subset of the cases (n = 32) were interrogated with 2083 microRNAs (miRNAs) to evaluate their use for pancreatic cyst risk stratification. RESULTS As independent PCF biomarkers in 32 cases with histologic diagnoses, three miRNAs performed significantly better than mutant KRAS, mutant GNAS, carcinoembryonic antigen (CEA), and serum carbohydrate antigen 19-9 (CA19-9) in discriminating high-risk from low-risk cysts. The three elevated miRNAs in combination with mutant KRAS, mutant GNAS, and serum CA19-9 displayed similar diagnostic performance (miR-4461: area under the curve [AUC], 0.950; 95% confidence interval [CI], 0.800-1; miR-6723-5p: AUC, 0.958; 95% CI, 0.850-1; miR-6755-3p: AUC, 0.942; 95% CI, 0.816-1) in discriminating high-risk from low-risk cysts, when compared to mutant KRAS, mutant GNAS, CEA, and serum CA19-9 (AUC, 0.950; 95% CI, 0.825-1). In the absence of CA19-9, the three-marker panel of KRAS, GNAS, and miRNAs showed marginally improved performance compared with KRAS, GNAS, and CEA, which highlights the potential utility of miRNAs as biomarkers in PCF analysis. CONCLUSIONS These findings demonstrate that a multiomics biomarker approach with elevated PCF miRNAs with mutant KRAS, mutant GNAS, and serum CA19-9 may help in better detecting high-risk cysts for early clinical intervention.
Collapse
Affiliation(s)
- Mohamed H Maher
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Warapen Treekitkarnmongkol
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sayak Ghatak
- Division of Pathology and Laboratory Medicine, Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jianliang Dai
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Suyu Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tristian Nguyen
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dzifa Y Duose
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael P Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tony Y Hu
- Center for Cellular and Molecular Diagnostics, Department of Cell and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Mark W Hurd
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Pamela L Paris
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| | - Kimberly S Kirkwood
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
- Section of Hepatopancreaticobiliary Surgery, Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, USA
| | - Anirban Maitra
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rajyalakshmi Luthra
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Pathology and Laboratory Medicine, Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Subrata Sen
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sinchita Roy-Chowdhuri
- Division of Pathology and Laboratory Medicine, Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Pathology and Laboratory Medicine, Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
8
|
Luchini C. Diagnostic Pearls and Pitfalls in the Evaluation of Biopsies of the Pancreas. Arch Pathol Lab Med 2025; 149:e54-e62. [PMID: 38387616 DOI: 10.5858/arpa.2023-0426-ra] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2023] [Indexed: 02/24/2024]
Abstract
CONTEXT.— The examination of small pancreatic biopsies is a difficult task for pathologists. This is due to the scant and fragmented material often obtained from diagnostic procedures as well as the significant overlap between different neoplastic and nonneoplastic entities. In the upcoming neoadjuvant era, biopsies could become even more important, representing the only possibility to look at the real histomorphology of tumors before chemotherapy-induced modifications. OBJECTIVES.— To summarize and discuss the state-of-the-art diagnostic workflow for small pancreatic biopsies, including the most important morphologic and immunohistochemical features and molecular alterations. The main diagnostic pearls and pitfalls of this challenging scenario are also discussed. The most important topics of this review are represented by: (1) pancreatic ductal adenocarcinoma, along with its main differential diagnoses, including autoimmune pancreatitis; (2) solid hypercellular neoplasms, including neuroendocrine neoplasms, acinar cell carcinoma, pancreatoblastoma, and solid pseudopapillary neoplasms; and (3) cystic lesions. Real-world considerations will also be presented and discussed. DATA SOURCES.— Sources included a literature review of published studies and the author's own work. CONCLUSIONS.— The correct diagnosis of pancreatic lesions is a crucial step in the therapeutic journey of patients. It should be based on robust, standardized, and reliable hallmarks. As presented and discussed here, the integration of morphology with immunohistochemistry, and, in selected cases, with molecular analysis, represents a decisive step in this complex scenario.
Collapse
Affiliation(s)
- Claudio Luchini
- From the Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy; and the ARC-Net Research Center, University of Verona, Verona, Italy
| |
Collapse
|
9
|
Afghani E, Lennon AM. What Is the Latest in Pancreatic Cysts? Gastroenterol Clin North Am 2025; 54:189-203. [PMID: 39880527 DOI: 10.1016/j.gtc.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Pancreatic cysts are common incidental findings. The understanding of pancreatic cysts has evolved tremendously over the past few decades. Molecular diagnostic and endoscopic techniques have led to more precise characterization of cyst types and interventions to improve patient outcomes. This article outlines these recent innovations in pancreatic cyst diagnosis and management.
Collapse
Affiliation(s)
- Elham Afghani
- Department of Medicine, Johns Hopkins University, 1830 East Monument Street, Room 436, Baltimore, MD 21287, USA
| | - Anne Marie Lennon
- Department of Medicine, University of Pittsburgh, 3550 Terrace Street, 1218 Scaife Hall, Pittsburgh, PA 15261, USA.
| |
Collapse
|
10
|
Kwan MC, Pitman MB, Zhang ML. Cytologic, histologic, and clinical correlation of minor mutations in pancreatic cysts. Cancer Cytopathol 2025; 133:e22935. [PMID: 39865498 DOI: 10.1002/cncy.22935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/19/2024] [Accepted: 12/05/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND Major mutations (e.g., KRAS, GNAS, TP53, SMAD4) in pancreatic cyst fluid (PCF) are useful for classifying and risk stratifying certain cyst types, particularly in cases with nondiagnostic cytology. However, the significance of uncommon minor mutations in PCF has yet to be reported. METHODS In total, 127 PCF specimens (2014-2021) from 121 patients that underwent molecular analysis were identified, and detailed clinicopathologic data were recorded. Molecular testing was performed using a laboratory-developed next-generation sequencing panel. RESULTS Forty-five variants other than KRAS, GNAS, RNF43, TP53, CDKN2A, and SMAD4 were detected. Variants that were detected in five or more cases included ARID1A (n = 28), VHL (n = 17), BRAF (n = 12), ATM (n = 8), APC (n = 8), MEN1 (n = 5), serine threonine kinase 11 (STK11; n = 5), PIK3CA (n = 5), and CDH1 (n = 5). Thirty-eight of 121 patients (31%) had histologic confirmation on follow-up resection. Twenty-seven of 28 cysts (96%) with ARID1A mutations had concurrent KRAS/GNAS mutations; 17 (61%) were diagnosed as neoplastic mucinous cysts on cytology, and 10 (36%) were diagnosed as intraductal papillary mucinous neoplasm (IPMN) on histology (80% low grade). No patients developed disease recurrence or died of disease. Cysts with STK11 mutations had RAS co-mutations (KRAS, n = 5; NRAS, n = 1), and four of those five cysts (80%) were mucinous neoplasms with high-grade atypia on cytology. All three resection specimens were IPMNs with high-grade dysplasia or invasive carcinoma, and two of those patients died of disease. CONCLUSIONS In PCFs, ARID1A mutations were consistently associated with IPMNs (predominantly low grade) with no recurrences or deaths from disease. STK11 mutations appeared to be associated with high-risk mucinous cysts. The detection of minor variants may provide useful preoperative information and add value beyond single-gene genotyping of major mutations.
Collapse
Affiliation(s)
- Melanie C Kwan
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Martha B Pitman
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - M Lisa Zhang
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Corradi C, Gentiluomo M, Adsay V, Sainz J, Camisa PR, Wlodarczyk B, Crippa S, Tavano F, Capurso G, Campa D. Multi-omic markers of intraductal papillary mucinous neoplasms progression into pancreatic cancer. Semin Cancer Biol 2025; 109:25-43. [PMID: 39733817 DOI: 10.1016/j.semcancer.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most lethal and common form of pancreatic cancer, it has no specific symptoms, and most of the patients are diagnosed when the disease is already at an advanced stage. Chemotherapy typically has only a modest effect, making surgery the most effective treatment option. However, only a small percentage of patients are amenable to surgery. One viable strategy to reduce PDAC death burden associated with the disease is to focus on precursor lesions and identify markers able to predict who will evolve into PDAC. While most PDACs are believed to be preceded by pancreatic intraepithelial neoplasms (PanINs), 5-10 % arise from Intraductal papillary mucinous neoplasms (IPMNs), which are mass-forming cystic lesions that are very common in the general population. IPMNs offer an invaluable model of pancreatic carcinogenesis for researchers to analyse, as well as a target population for PDAC early detection by clinicians. The evolution of IPMN into cancer is a complex and multistep process, therefore the identification of individual markers will not be the solution. In recent years, multiple omics technologies have been instrumental to identify possible biomarkers of IPMN progression and carcinogenesis. The only foreseeable strategy will be to integrate multi-omics data, alongside clinical and morphological features, into a progression score or signature using either standard epidemiologic tools or artificial intelligence. The aim of this manuscript is to review the current knowledge on genetic biomarkers and to briefly mention also additional omics, such as metabolomics, the exposome, the miRNome and epigenomics of IPMNs.
Collapse
Affiliation(s)
| | | | - Volkan Adsay
- Department of Pathology, Koç University School of Medicine and Koç University Research Center for Translational Medicine, Istanbul, Turkey
| | - Juan Sainz
- Department of Biochemistry and Molecular Biology, University of Granada, Granada, Spain
| | - Paolo Riccardo Camisa
- Division of Pancreatic Surgery and Transplantation, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Barbara Wlodarczyk
- Department of Digestive Tract Diseases, Medical University of Lodz, Lodz, Poland
| | - Stefano Crippa
- Division of Pancreatic Surgery and Transplantation, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Francesca Tavano
- Division of Gastroenterology and Research Laboratory, Fondazione IRCCS "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, Italy
| | - Gabriele Capurso
- Vita-Salute San Raffaele University, Milan, Italy; Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy.
| |
Collapse
|
12
|
Hamada T, Oyama H, Nevo D, Tange S, Takaoka S, Kawaguchi Y, Ishigaki K, Noguchi K, Saito T, Sato T, Suzuki T, Takahara N, Tanaka M, Hasegawa K, Ushiku T, Nakai Y, Petrov MS, Fujishiro M. Risk factors for pancreatic cancer in individuals with intraductal papillary mucinous neoplasms and no high-risk stigmata during up to 5 years of surveillance: a prospective longitudinal cohort study. Gut 2025:gutjnl-2024-333259. [PMID: 39870394 DOI: 10.1136/gutjnl-2024-333259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 01/11/2025] [Indexed: 01/29/2025]
Abstract
BACKGROUND Cyst size, its growth rate, and diameter of the main pancreatic duct (MPD) are all associated with pancreatic carcinoma prevalence in intraductal papillary mucinous neoplasms (IPMNs). OBJECTIVE To examine the above factors in relation to future risk of incident pancreatic carcinoma in individuals with IPMNs harbouring no high-risk stigmata. DESIGN In a prospective longitudinal cohort, we analysed 2549 patients with IPMNs. A multivariable cause-specific Cox proportional hazards regression model was built to estimate HRs for incident pancreatic carcinoma. RESULTS IPMN size at baseline and its annual growth rate over 2 years of follow-up were associated with incident pancreatic carcinoma (ptrend<0.001). The multivariable cause-specific HR per 10 mm increase in IPMN size was 1.28 (95% CI 1.10 to 1.50). The annual growth rates of 1.5-2.4 mm/year and ≥2.5 mm/year over 2 years were associated with multivariable cause-specific HRs of 1.91 (95% CI 0.78 to 4.67) and 4.52 (95% CI 2.28 to 8.98), respectively (vs <1.5 mm/year). Neither IPMN size at 5 years nor its maximum growth rate during 5 years was associated with incident pancreatic carcinoma (ptrend>0.07). MPD diameter at 5 years was associated with incident pancreatic carcinoma (multivariable cause-specific HR per 2 mm increase, 2.12; 95% CI 1.72 to 2.63). A predictive nomogram was generated for calculating the risk of incident pancreatic carcinoma. CONCLUSION IPMN size and its growth rate predict future pancreatic carcinoma risk only during first 5 years of follow-up. MPD diameter at 5 years may identify patients who still harbour a high risk for pancreatic carcinoma.
Collapse
Affiliation(s)
- Tsuyoshi Hamada
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Hepato-Biliary-Pancreatic Medicine, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroki Oyama
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Daniel Nevo
- Department of Statistics and Operations Research, Tel Aviv University, Tel Aviv, Israel
| | - Shuichi Tange
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinya Takaoka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Endoscopy and Endoscopic Surgery, The University of Tokyo Hospital, Tokyo, Japan
| | - Yoshikuni Kawaguchi
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazunaga Ishigaki
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kensaku Noguchi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomotaka Saito
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tatsuya Sato
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tatsunori Suzuki
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naminatsu Takahara
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mariko Tanaka
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Hasegawa
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yousuke Nakai
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Internal Medicine, Institute of Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - Maxim S Petrov
- School of Medicine, University of Auckland, Auckland, New Zealand
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
13
|
Jamali Z, Razipour M, Zargar M, Ghasemnejad-Berenji H, Akrami SM. Ovarian cancer extracellular vesicle biomarkers. Clin Chim Acta 2025; 565:120011. [PMID: 39437983 DOI: 10.1016/j.cca.2024.120011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Ovarian cancer (OC) remains a significant women's health concern due to its high mortality rate and the challenges posed by late detection. Exploring novel biomarkers could lead to earlier, more specific diagnoses and improved survival rates for OC patients. This review focuses on biomarkers associated with extracellular vesicles (EVs) found in various proximal fluids, including urine, ascites, utero-tubal lavage fluid of OC patients. We highlight these proximal fluids as rich sources of potential biomarkers. The review explains the roles of EV biomarkers in ovarian cancer progression and discusses EV-related proteins and miRNAs as potential diagnostic or prognostic indicators and therapeutic targets. Finally, we highlighted the limitations of examining proximal fluids as sources of biomarkers and encourage researchers to proactively pursue innovative solutions to overcome these challenges.
Collapse
Affiliation(s)
- Zeinab Jamali
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Razipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Zargar
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hojat Ghasemnejad-Berenji
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Seyed Mohammad Akrami
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Dhar J, Samanta J, Nabi Z, Aggarwal M, Conti Bellocchi MC, Facciorusso A, Frulloni L, Crinò SF. Endoscopic Ultrasound-Guided Pancreatic Tissue Sampling: Lesion Assessment, Needles, and Techniques. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:2021. [PMID: 39768901 PMCID: PMC11727853 DOI: 10.3390/medicina60122021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/15/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025]
Abstract
Endoscopic ultrasound (EUS)-guided tissue sampling includes the techniques of fine needle aspiration (FNA) and fine needle biopsy (FNB), and both procedures have revolutionized specimen collection from the gastrointestinal tract, especially from remote/inaccessible organs. EUS-FNB has replaced FNA as the procedure of choice for tissue acquisition in solid pancreatic lesions (SPLs) across various society guidelines. FNB specimens provide a larger histological tissue core (preserving tissue architecture) with fewer needle passes, and this is extremely relevant in today's era of precision and personalized molecular medicine. Innovations in needle tip design are constantly under development to maximize diagnostic accuracy by enhancing histological sampling capabilities. But, apart from the basic framework of the needle, various other factors play a role that influence diagnostic outcomes, namely, sampling techniques (fanning, aspiration or suction, and number of passes), collection methods, on-site evaluation (rapid, macroscopic, or visual), and specimen processing. The choice taken depends strongly on the endoscopist's preference, available resources at the disposal, and procedure objectives. Hence, in this review, we explicate in detail the concepts and available literature at our disposal on the topic of EUS-guided pancreatic tissue sampling to best guide any practicing gastroenterologist/endoscopist in a not-to-ideal set-up, which EUS-guided tissue acquisition technique is the "best" for their case to augment their diagnostic outcomes.
Collapse
Affiliation(s)
- Jahnvi Dhar
- Department of Gastroenterology, Adesh Medical College and Hospital, Kurukshetra 136134, India;
| | - Jayanta Samanta
- Department of Gastroenterology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India;
| | - Zaheer Nabi
- Department of Gastroenterology, Asian Institute of Gastroenterology, Hyderabad 500082, India;
| | - Manik Aggarwal
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Maria Cristina Conti Bellocchi
- Department of Medicine, Diagnostic and Interventional Endoscopy of the Pancreas, The Pancreas Institute, University Hospital of Verona, 37134 Verona, Italy; (M.C.C.B.); (L.F.)
| | - Antonio Facciorusso
- Gastroenterology Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
- Clinical Effectiveness Research Group, Faculty of Medicine, Institute of Health and Society, University of Oslo, 0372 Oslo, Norway
| | - Luca Frulloni
- Department of Medicine, Diagnostic and Interventional Endoscopy of the Pancreas, The Pancreas Institute, University Hospital of Verona, 37134 Verona, Italy; (M.C.C.B.); (L.F.)
| | - Stefano Francesco Crinò
- Department of Medicine, Diagnostic and Interventional Endoscopy of the Pancreas, The Pancreas Institute, University Hospital of Verona, 37134 Verona, Italy; (M.C.C.B.); (L.F.)
| |
Collapse
|
15
|
Smilkou S, Kaklamanis L, Balgouranidou I, Linardou H, Papatheodoridi AM, Zagouri F, Razis E, Kakolyris S, Psyrri A, Papadimitriou C, Markou A, Lianidou E. Direct comparison of an ultrasensitive real-time PCR assay with droplet digital PCR for the detection of PIK3CA hotspot mutations in primary tumors, plasma cell-free DNA and paired CTC-derived gDNAs. Front Oncol 2024; 14:1435559. [PMID: 39711963 PMCID: PMC11659196 DOI: 10.3389/fonc.2024.1435559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/28/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction Detection of PIK3CA mutations in primary tumors and liquid biopsy samples is of increasing importance for treatment decisions and therapy resistance in many types of cancer. The aim of the present study was to directly compare the efficacy of a relatively inexpensive ultrasensitive real-time PCR with the well-established and highly sensitive technology of ddPCR for the detection of the three most common hotspot mutations of PIK3CA, in exons 9 and 20, that are all of clinical importance in various types of cancer. Patients and methods We analyzed 42 gDNAs from primary tumors (FFPEs), 29 plasma-cfDNA samples, and 29 paired CTC-derived gDNAs, all from patients with ER+ metastatic breast cancer, and plasma from 10 healthy donors. The same blood draws were used for CTC isolation using EpCAM beads for positive immunomagnetic enrichment. All FFPEs and plasma-cfDNA samples were analyzed in parallel for PIK3CA mutations by ultrasensitive real-time PCR assay and droplet digital PCR. Results In gDNAs from FFPEs, using ultrasensitive real-time PCR, the p.E545K mutation was detected in 22/42(52.4%), and the p.E542K and p.H1047R mutations were detected in 14/42(33.3%) and 16/42(38.1%), respectively. Using ddPCR, the p.E545K mutation was detected in 22/42(52.4%), p.E542K in 17/42(40.5%), and p.H1047R in 19/42(45.2%) samples, revealing a concordance between the two methodologies of 81%, 78.6% and 78.6% for each mutation respectively. In plasma-cfDNA, using ultrasensitive real-time PCR, the p.E545K mutation was detected in 11/29(38%) and both p.E542K and p.H1047R mutations in 2/29(6.9%).In the same plasma-cfDNA samples using ddPCR, p.E545K was detected in 1/29(3.5%), p.E542K in 2/29(6.9%), and p.H1047R in 3/29(10.5%) samples, revealing a concordance of 65.5%,100% and 93.1% for each mutation respectively. In paired CTC-derived gDNAs p.E545K was detected in 11/29(38%), p.E542K in 3/29(10.3%), and p.H1047R in 7/29(24.1%) samples. Conclusions This low-cost, high-throughput and ultrasensitive real-time PCR assay provides accurate and specific detection of PIK3CA hotspot mutations in liquid biopsy samples and gives similar results to ddPCR. This assay can be performed in labs where digital PCR instrumentation is not available. In CTC-derived gDNA and paired plasma-cfDNA, PIK3CA mutations detected were not identical, revealing that CTC and plasma-cfDNA give complementary information.
Collapse
Affiliation(s)
- Stavroula Smilkou
- Analysis of Circulating Tumor Cells, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, Athens, Greece
| | - Loukas Kaklamanis
- Department of Pathology, Onassis Cardiac Surgery Center, Athens, Greece
| | - Ioanna Balgouranidou
- Department of Medical Oncology, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | | | - Alkistis Maria Papatheodoridi
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Flora Zagouri
- Department of Clinical Therapeutics, School of Medicine, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelia Razis
- Third Department of Medical Oncology, Hygeia Hospital, Athens, Greece
| | - Stylianos Kakolyris
- Department of Medical Oncology, University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Amanda Psyrri
- Section of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Attikon University Hospital, Athens, Greece
| | - Christos Papadimitriou
- Oncology Unit, 2nd Department of Surgery, School of Medicine, Aretaieio Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athina Markou
- Analysis of Circulating Tumor Cells, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, Athens, Greece
| | - Evi Lianidou
- Analysis of Circulating Tumor Cells, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, Athens, Greece
| |
Collapse
|
16
|
Millastre J, Hermoso-Durán S, de Solórzano MO, Fraunhoffer N, García-Rayado G, Vega S, Bujanda L, Sostres C, Lanas Á, Velázquez-Campoy A, Abian O. Thermal Liquid Biopsy: A Promising Tool for the Differential Diagnosis of Pancreatic Cystic Lesions and Malignancy Detection. Cancers (Basel) 2024; 16:4024. [PMID: 39682210 PMCID: PMC11640424 DOI: 10.3390/cancers16234024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Pancreatic cystic lesions (PCLs) are a heterogeneous group of lesions with increasing incidence, usually identified incidentally on imaging studies (multidetector computed tomography (MDCT), magnetic resonance imaging (MRI), or endoscopic ultrasound (EUS)) [...].
Collapse
Affiliation(s)
- Judith Millastre
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain; (J.M.); (M.O.d.S.); (G.G.-R.); (C.S.); (Á.L.)
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Sonia Hermoso-Durán
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain;
| | - María Ortiz de Solórzano
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain; (J.M.); (M.O.d.S.); (G.G.-R.); (C.S.); (Á.L.)
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Nicolas Fraunhoffer
- Programa Franco-Argentino de Estudio del Cáncer de Páncreas, Buenos Aires, Argentina;
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, CNRS UMR, Aix-Marseille Université, 13009 Marseille, France
- Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Equipe Labellisée La Ligue, 13288 Marseille, France
| | - Guillermo García-Rayado
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain; (J.M.); (M.O.d.S.); (G.G.-R.); (C.S.); (Á.L.)
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Sonia Vega
- Institute for Biocomputation and Physics of Complex Systems, University of Zaragoza, 50018 Zaragoza, Spain;
| | - Luis Bujanda
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain;
- Donostia University Hospital, University of the Basque Country (UPV/EHU), 20014 San Sebastian, Spain
| | - Carlos Sostres
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain; (J.M.); (M.O.d.S.); (G.G.-R.); (C.S.); (Á.L.)
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Ángel Lanas
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain; (J.M.); (M.O.d.S.); (G.G.-R.); (C.S.); (Á.L.)
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Adrián Velázquez-Campoy
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain;
- Institute for Biocomputation and Physics of Complex Systems, University of Zaragoza, 50018 Zaragoza, Spain;
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, 50009 Zaragoza, Spain
| | - Olga Abian
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain;
- Institute for Biocomputation and Physics of Complex Systems, University of Zaragoza, 50018 Zaragoza, Spain;
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, 50009 Zaragoza, Spain
| |
Collapse
|
17
|
Iyer MK, Fletcher A, Shi C, Chen F, Kanu E, Eckhoff AM, Bao M, Frankel TL, Chinnaiyan AM, Nussbaum DP, Allen PJ. Spatial Transcriptomics of IPMN Reveals Divergent Indolent and Malignant Lineages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620810. [PMID: 39554015 PMCID: PMC11565728 DOI: 10.1101/2024.10.29.620810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Purpose Intraductal papillary mucinous neoplasms (IPMN) occur in 5-10% of the population, but only a small minority progress to pancreatic ductal adenocarcinoma (PDAC). The lack of accurate predictors of high-risk disease leads both to unnecessary operations for indolent neoplasms as well as missed diagnoses of PDAC. Digital spatial RNA profiling (DSP-RNA) provides an opportunity to define and associate transcriptomic states with cancer risk. Experimental Design Whole-transcriptome DSP-RNA profiling was performed on 10 IPMN specimens encompassing the spectrum of dysplastic changes from normal duct to cancer. Ductal epithelial regions within each tissue were annotated as normal duct (NL), low-grade dysplasia (LGD), high-grade dysplasia (HGD), or invasive carcinoma (INV). Gene expression count data was generated by Illumina sequencing and analyzed with R/Bioconductor. Results Dimension reduction analysis exposed three clusters reflecting IPMN transcriptomic states denoted "normal-like" ( cNL ), "low-risk" ( cLR ) and "high-risk" ( cHR ). In addition to specific marker genes, the three states exhibited significant enrichment for the exocrine, classical, and basal-like programs in PDAC. Specifically, exocrine function diminished in cHR , classical activation distinguished neoplasia from cNL , and basal-like genes were specifically upregulated in cHR . Intriguingly, markers of cHR were detected in NL and LGD regions from specimens with PDAC but not low-grade IPMN. Conclusions DSP-RNA of IPMN revealed low-risk (indolent) and high-risk (malignant) expression programs that correlated with the activity of exocrine and basal-like PDAC signatures, respectively, and distinguished pathologically low-grade from malignant specimens. These findings contextualize IPMN pathogenesis and have the potential to transform existing risk stratification models. Statement of translational relevance Current consensus guidelines for management of intraductal papillary mucinous neoplasms (IPMN) of the pancreas utilize clinical and radiographic criteria for risk stratification. Unfortunately, the estimated positive predictive value of these criteria for IPMN-associated pancreatic ductal adenocarcinoma (PDAC) is under 50%, indicating that over half of pancreatectomies are performed for benign disease. Moreover, nearly 15% of patients who were deemed "low risk" by the same criteria harbored PDAC. Surgical resection of IPMN has maximal benefit when performed prior to the development of PDAC, as evidence of carcinoma has been associated with a high rate of recurrence and poor overall survival. Thus, the development of molecular diagnostics that improve the accuracy of IPMN risk classification would have immediate relevance for patient care, both in terms of better selecting patients for potentially curative operations, as well as sparing patients with low-risk lesions from invasive procedures.
Collapse
|
18
|
Lavista Ferres JM, Oviedo F, Robinson C, Chu L, Kawamoto S, Afghani E, He J, Klein AP, Goggins M, Wolfgang CL, Javed AA, Dodhia R, Papadopolous N, Kinzler K, Hruban RH, Weeks WB, Fishman EK, Lennon AM. Performance of explainable artificial intelligence in guiding the management of patients with a pancreatic cyst. Pancreatology 2024; 24:1182-1191. [PMID: 39261223 DOI: 10.1016/j.pan.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/12/2024] [Accepted: 09/01/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND/OBJECTIVES Pancreatic cyst management can be distilled into three separate pathways - discharge, monitoring or surgery- based on the risk of malignant transformation. This study compares the performance of artificial intelligence (AI) models to clinical care for this task. METHODS Two explainable boosting machine (EBM) models were developed and evaluated using clinical features only, or clinical features and cyst fluid molecular markers (CFMM) using a publicly available dataset, consisting of 850 cases (median age 64; 65 % female) with independent training (429 cases) and holdout test cohorts (421 cases). There were 137 cysts with no malignant potential, 114 malignant cysts, and 599 IPMNs and MCNs. RESULTS The EBM and EBM with CFMM models had higher accuracy for identifying patients requiring monitoring (0.88 and 0.82) and surgery (0.66 and 0.82) respectively compared with current clinical care (0.62 and 0.58). For discharge, the EBM with CFMM model had a higher accuracy (0.91) than either the EBM model (0.84) or current clinical care (0.86). In the cohort of patients who underwent surgical resection, use of the EBM-CFMM model would have decreased the number of unnecessary surgeries by 59 % (n = 92), increased correct surgeries by 7.5 % (n = 11), identified patients who require monitoring by 122 % (n = 76), and increased the number of patients correctly classified for discharge by 138 % (n = 18) compared to clinical care. CONCLUSIONS EBM models had greater sensitivity and specificity for identifying the correct management compared with either clinical management or previous AI models. The model predictions are demonstrated to be interpretable by clinicians.
Collapse
Affiliation(s)
| | - Felipe Oviedo
- AI for Good Lab, Microsoft Corporation, 4330 150th Ave NE, Redmond, WA, 98052, USA
| | - Caleb Robinson
- AI for Good Lab, Microsoft Corporation, 4330 150th Ave NE, Redmond, WA, 98052, USA
| | - Linda Chu
- Department of Radiology, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA
| | - Satomi Kawamoto
- Department of Radiology, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA
| | - Elham Afghani
- Department of Medicine, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA
| | - Jin He
- Department of Surgery, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA
| | - Alison P Klein
- Department of Medicine, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA; Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA; The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA
| | - Mike Goggins
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA
| | - Christopher L Wolfgang
- Department of Surgery, New York University School of Medicine, NYU, New York, NY, 10016, USA
| | - Ammar A Javed
- Department of Surgery, New York University School of Medicine, NYU, New York, NY, 10016, USA
| | - Rahul Dodhia
- AI for Good Lab, Microsoft Corporation, 4330 150th Ave NE, Redmond, WA, 98052, USA
| | - Nick Papadopolous
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA; Ludwig Center and Howard Hughes Medical Institute at the Sidney Kimmel Cancer Center, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA
| | - Ken Kinzler
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA; Ludwig Center and Howard Hughes Medical Institute at the Sidney Kimmel Cancer Center, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA
| | - Ralph H Hruban
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA
| | - William B Weeks
- AI for Good Lab, Microsoft Corporation, 4330 150th Ave NE, Redmond, WA, 98052, USA
| | - Elliot K Fishman
- Department of Radiology, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA
| | - Anne Marie Lennon
- Department of Radiology, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA; Department of Medicine, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA; Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA; Ludwig Center and Howard Hughes Medical Institute at the Sidney Kimmel Cancer Center, Johns Hopkins University, 1800 Orleans Street, Baltimore, MD, 21287, USA.
| |
Collapse
|
19
|
Pollini T, Todeschini L, Maker AV. Pancreas Cyst Diagnosis and Advances in Cyst Fluid Analysis. Surg Clin North Am 2024; 104:965-974. [PMID: 39237171 DOI: 10.1016/j.suc.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Pancreatic Cystic Neoplasms (PCN) represent a diverse group of tumors, some of which may progress to pancreatic cancer. Considering their high prevalence in the general population, the development of reliable biomarkers is crucial. The ideal biomarker will accurately diagnose the subtype of PCN and assess the risk of high-grade dysplasia or invasive cancer. Cyst fluid analysis has emerged as a promising approach to accomplish this goal, yet no single marker has yet gained unanimous support for routine inclusion in PCN evaluation.
Collapse
Affiliation(s)
- Tommaso Pollini
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco
| | - Letizia Todeschini
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco
| | - Ajay V Maker
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco.
| |
Collapse
|
20
|
Gonda TA, Cahen DL, Farrell JJ. Pancreatic Cysts. N Engl J Med 2024; 391:832-843. [PMID: 39231345 DOI: 10.1056/nejmra2309041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Affiliation(s)
- Tamas A Gonda
- From the Division of Gastroenterology and Hepatology, Department of Medicine, New York University (NYU) Grossman School of Medicine and NYU Langone Health, New York (T.A.G.); the Division of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands (D.L.C); and the Division of Digestive Diseases, Department of Medicine, Yale University School of Medicine and Yale New Haven Health, New Haven, CT (J.J.F.)
| | - Djuna L Cahen
- From the Division of Gastroenterology and Hepatology, Department of Medicine, New York University (NYU) Grossman School of Medicine and NYU Langone Health, New York (T.A.G.); the Division of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands (D.L.C); and the Division of Digestive Diseases, Department of Medicine, Yale University School of Medicine and Yale New Haven Health, New Haven, CT (J.J.F.)
| | - James J Farrell
- From the Division of Gastroenterology and Hepatology, Department of Medicine, New York University (NYU) Grossman School of Medicine and NYU Langone Health, New York (T.A.G.); the Division of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands (D.L.C); and the Division of Digestive Diseases, Department of Medicine, Yale University School of Medicine and Yale New Haven Health, New Haven, CT (J.J.F.)
| |
Collapse
|
21
|
Thompson ED. Neoplastic Progression in Macroscopic Precursor Lesions of the Pancreas. Arch Pathol Lab Med 2024; 148:980-988. [PMID: 38386006 DOI: 10.5858/arpa.2023-0358-ra] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 02/23/2024]
Abstract
CONTEXT.— Macroscopic precursor lesions of the pancreas represent a complex clinical management problem. Molecular characterization of pancreatic cysts has helped to confirm and refine clinical and pathologic classifications of these lesions, inform our understanding of tumorigenesis in the pancreas, and provide opportunities for preoperative diagnosis. OBJECTIVE.— To review the pathologic classification of macroscopic cystic lesions of the pancreas: intraductal papillary mucinous neoplasms (IPMNs), mucinous cystic neoplasms (MCNs), intraductal oncocytic papillary neoplasms (IOPNs), and intraductal tubulopapillary neoplasms (ITPNs), and to describe our current state of understanding of their molecular underpinnings, relationship to invasive carcinomas, and implications for diagnosis and prognostication. DATA SOURCES.— We assessed the current primary literature and current World Health Organization Classification of Digestive System Tumours. CONCLUSIONS.— Macroscopic cystic lesions of the pancreas are morphologically and molecularly diverse. IPMNs and MCNs share mucinous cytoplasm with papillae. MCNs are defined by ovarian-type stroma. IOPNs have granular eosinophilic cytoplasm, prominent nucleoli, and complex, arborizing papillae. ITPNs demonstrate complex, back-to-back tubules and anastomosing papillae and lack prominent intracellular mucin. IPMNs and MCNs are characterized by driver mutations in KRAS/GNAS (IPMNs) and KRAS (MCNs), with later driver events in RNF43, CDKN2A, SMAD4, and TP53. In contrast, IOPNs and ITPNs have recurrent rearrangements in PRKACA/PRKACB and MAPK-associated genes, respectively. The recurrent alterations described in cysts provide an opportunity for diagnosis using aspirated cyst fluid. Molecular characterization of IPMNs shows a striking spatial and mutational heterogeneity, challenging traditional models of neoplastic development and creating challenges to interpretation of cyst fluid sequencing results.
Collapse
Affiliation(s)
- Elizabeth D Thompson
- From the Departments of Pathology and Oncology, Sol Goldman Pancreatic Cancer Research Center, and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
22
|
Patel V, Abdelbaki A, Thosani NC, Krishna SG. Endoscopic ultrasound-guided radiofrequency ablation of pancreatic tumors. Curr Opin Gastroenterol 2024; 40:369-378. [PMID: 38662451 DOI: 10.1097/mog.0000000000001026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/09/2024]
Abstract
PURPOSE OF REVIEW Surgery is a cornerstone in the management of pancreatic cancer and precancerous pancreatic lesions. However, many patients are not suitable candidates for surgery at the time of diagnosis for various reasons. Endoscopic ultrasound-guided radiofrequency ablation (EUS-RFA) appears to be a promising treatment option for patients who are ineligible for surgery for management of pancreatic adenocarcinoma (PDAC), and pancreatic neuroendocrine tumors (PNETs), and pancreatic cystic lesions (PCLs). RECENT FINDINGS EUS-RFA may serve as an adjunct to chemotherapy or palliative measures for inoperable cases of PDAC. Given its feasibility and efficacy, EUS-RFA has an evolving niche as a minimally invasive and potentially definitive treatment for PNETs and high-risk PCLs such as intraductal papillary mucinous neoplasms (IPMNs). EUS-RFA is a generally well tolerated procedure, with abdominal pain and acute pancreatitis being the most common adverse effects, though the risk can be mitigated through prophylactic measures. SUMMARY There is an increasing body of evidence to support the use of EUS-RFA in managing pancreatic lesions, either as definitive, adjunctive, or palliative treatment, depending on lesion type.
Collapse
Affiliation(s)
- Vanisha Patel
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Ohio
| | - Ahmed Abdelbaki
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Ohio
| | - Nirav C Thosani
- Center for Interventional Gastroenterology at UTHealth (iGUT), Division of Elective General Surgery, Department of Surgery, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Somashekar G Krishna
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Ohio
| |
Collapse
|
23
|
Rogowska J, Semeradt J, Durko Ł, Małecka-Wojciesko E. Diagnostics and Management of Pancreatic Cystic Lesions-New Techniques and Guidelines. J Clin Med 2024; 13:4644. [PMID: 39200786 PMCID: PMC11355509 DOI: 10.3390/jcm13164644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Pancreatic cystic lesions (PCLs) are increasingly diagnosed owing to the wide use of cross-sectional imaging techniques. Accurate identification of PCL categories is critical for determining the indications for surgical intervention or surveillance. The classification and management of PCLs rely on a comprehensive and interdisciplinary evaluation, integrating clinical data, imaging findings, and cyst fluid markers. EUS (endoscopic ultrasound) has become the widely used diagnostic tool for the differentiation of pancreatic cystic lesions, offering detailed evaluation of even small pancreatic lesions with high sensitivity and specificity. Additionally, endoscopic ultrasound-fine-needle aspiration enhances diagnostic capabilities through cytological analysis and the assessment of fluid viscosity, tumor glycoprotein concentration, amylase levels, and molecular scrutiny. These detailed insights play a pivotal role in improving the clinical prognosis and management of pancreatic neoplasms. This review will focus mainly on the latest recommendations for the differentiation, management, and treatment of pancreatic cystic lesions, highlighting their clinical significance.
Collapse
Affiliation(s)
- Jagoda Rogowska
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-647 Lodz, Poland; (J.S.); (Ł.D.); (E.M.-W.)
| | | | | | | |
Collapse
|
24
|
Bogdanski AM, van Hooft JE, Boekestijn B, Bonsing BA, Wasser MNJM, Klatte DCF, van Leerdam ME. Aspects and outcomes of surveillance for individuals at high-risk of pancreatic cancer. Fam Cancer 2024; 23:323-339. [PMID: 38619782 PMCID: PMC11255004 DOI: 10.1007/s10689-024-00368-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/24/2024] [Indexed: 04/16/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related deaths and is associated with a poor prognosis. The majority of these cancers are detected at a late stage, contributing to the bad prognosis. This underscores the need for novel, enhanced early detection strategies to improve the outcomes. While population-based screening is not recommended due to the relatively low incidence of PDAC, surveillance is recommended for individuals at high risk for PDAC due to their increased incidence of the disease. However, the outcomes of pancreatic cancer surveillance in high-risk individuals are not sorted out yet. In this review, we will address the identification of individuals at high risk for PDAC, discuss the objectives and targets of surveillance, outline how surveillance programs are organized, summarize the outcomes of high-risk individuals undergoing pancreatic cancer surveillance, and conclude with a future perspective on pancreatic cancer surveillance and novel developments.
Collapse
Affiliation(s)
- Aleksander M Bogdanski
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Jeanin E van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Bas Boekestijn
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin N J M Wasser
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Derk C F Klatte
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Wang Y, Lih TM, Lee JW, Ohtsuka T, Hozaka Y, Mino-Kenudson M, Adsay NV, Luchini C, Scarpa A, Maker AV, Kim GE, Paulino J, Chen L, Jiao L, Sun Z, Goodman D, Pflüger MJ, Roberts NJ, Matthaei H, Wood LD, Furukawa T, Zhang H, Hruban RH. Multi-omic profiling of intraductal papillary neoplasms of the pancreas reveals distinct expression patterns and potential markers of progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.07.602385. [PMID: 39005476 PMCID: PMC11245086 DOI: 10.1101/2024.07.07.602385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
In order to advance our understanding of precancers in the pancreas, 69 pancreatic intraductal papillary neoplasms (IPNs), including 64 intraductal papillary mucinous neoplasms (IPMNs) and 5 intraductal oncocytic papillary neoplasms (IOPNs), 32 pancreatic cyst fluid samples, 104 invasive pancreatic ductal adenocarcinomas (PDACs), 43 normal adjacent tissues (NATs), and 76 macro-dissected normal pancreatic ducts (NDs) were analyzed by mass spectrometry. A total of 10,246 proteins and 22,284 glycopeptides were identified in all tissue samples, and 756 proteins with more than 1.5-fold increase in abundance in IPMNs relative to NDs were identified, 45% of which were also identified in cyst fluids. The over-expression of selected proteins was validated by immunolabeling. Proteins and glycoproteins overexpressed in IPMNs included those involved in glycan biosynthesis and the immune system. In addition, multiomics clustering identified two subtypes of IPMNs. This study provides a foundation for understanding tumor progression and targets for earlier detection and therapies. Significance This multilevel characterization of intraductal papillary neoplasms of the pancreas provides a foundation for understanding the changes in protein and glycoprotein expression during the progression from normal duct to intraductal papillary neoplasm, and to invasive pancreatic carcinoma, providing a foundation for informed approaches to earlier detection and treatment.
Collapse
|
26
|
Krishna S, Abdelbaki A, Hart PA, Machicado JD. Endoscopic Ultrasound-Guided Needle-Based Confocal Endomicroscopy as a Diagnostic Imaging Biomarker for Intraductal Papillary Mucinous Neoplasms. Cancers (Basel) 2024; 16:1238. [PMID: 38539568 PMCID: PMC10969577 DOI: 10.3390/cancers16061238] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 11/11/2024] Open
Abstract
Pancreatic cancer is on track to become the second leading cause of cancer-related deaths by 2030, yet there is a lack of accurate diagnostic tests for early detection. Intraductal papillary mucinous neoplasms (IPMNs) are precursors to pancreatic cancer and are increasingly being detected. Despite the development and refinement of multiple guidelines, diagnosing high-grade dysplasia or cancer in IPMNs using clinical, radiologic, endosonographic, and cyst fluid features still falls short in terms of accuracy, leading to both under- and overtreatment. EUS-guided needle-based confocal laser endomicroscopy (nCLE) is a novel technology that allows real-time optical biopsies of pancreatic cystic lesions. Emerging data has demonstrated that EUS-nCLE can diagnose and risk stratify IPMNs more accurately than conventional diagnostic tools. Implementing EUS-nCLE in clinical practice can potentially improve early diagnosis of pancreatic cancer, reduce unnecessary surgeries of IPMNs with low-grade dysplasia, and advance the field of digital pathomics. In this review, we summarize the current evidence that supports using EUS-nCLE as a diagnostic imaging biomarker for diagnosing IPMNs and for risk stratifying their degree of neoplasia. Moreover, we will present emerging data on the role of adding artificial intelligence (AI) algorithms to nCLE and integrating novel fluid biomarkers into nCLE.
Collapse
Affiliation(s)
- Shreyas Krishna
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (S.K.); (A.A.)
| | - Ahmed Abdelbaki
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (S.K.); (A.A.)
| | - Phil A. Hart
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (S.K.); (A.A.)
| | - Jorge D. Machicado
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
27
|
Hu Y, Jones D, Esnakula AK, Krishna SG, Chen W. Molecular Pathology of Pancreatic Cystic Lesions with a Focus on Malignant Progression. Cancers (Basel) 2024; 16:1183. [PMID: 38539517 PMCID: PMC10969285 DOI: 10.3390/cancers16061183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 11/11/2024] Open
Abstract
The malignant progression of pancreatic cystic lesions (PCLs) remains understudied with a knowledge gap, yet its exploration is pivotal for effectively stratifying patient risk and detecting cancer at its earliest stages. Within this review, we delve into the latest discoveries on the molecular level, revealing insights into the IPMN molecular landscape and revised progression model, associated histologic subtypes, and the role of inflammation in the pathogenesis and malignant progression of IPMN. Low-grade PCLs, particularly IPMNs, can develop into high-grade lesions or invasive carcinoma, underscoring the need for long-term surveillance of these lesions if they are not resected. Although KRAS and GNAS remain the primary oncogenic drivers of neoplastic development in IPMNs, additional genes that are important in tumorigenesis have been recently identified by whole exome sequencing. A more complete understanding of the genes involved in the molecular progression of IPMN is critical for effective monitoring to minimize the risk of malignant progression. Complicating these strategies, IPMNs are also frequently multifocal and multiclonal, as demonstrated by comparative molecular analysis. Algorithms for preoperative cyst sampling and improved radiomic techniques are emerging to model this spatial and temporal genetic heterogeneity better. Here, we review the molecular pathology of PCLs, focusing on changes associated with malignant progression. Developing models of molecular risk stratification in PCLs which can complement radiologic and clinical features, facilitate the early detection of pancreatic cancer, and enable the development of more personalized surveillance and management strategies are summarized.
Collapse
Affiliation(s)
- Yan Hu
- James Molecular Laboratory, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (Y.H.); (D.J.)
| | - Dan Jones
- James Molecular Laboratory, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (Y.H.); (D.J.)
| | - Ashwini K. Esnakula
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Somashekar G. Krishna
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Wei Chen
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| |
Collapse
|
28
|
Kwan MC, Pitman MB, Fernandez-Del Castillo C, Zhang ML. Revisiting the performance of cyst fluid carcinoembryonic antigen as a diagnostic marker for pancreatic mucinous cysts: a comprehensive 20-year institutional review. Gut 2024; 73:629-638. [PMID: 38195219 DOI: 10.1136/gutjnl-2023-331138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/27/2023] [Indexed: 01/11/2024]
Abstract
OBJECTIVE Elevated pancreatic cyst fluid carcinoembryonic antigen (CEA) has been routinely used to classify mucinous cysts. This study incorporates original data that established the CEA ≥192 ng/mL threshold with over 20 years of additional data and reassesses the diagnostic performance of CEA for differentiating mucinous from non-mucinous cysts. DESIGN 1169 pancreatic cysts (1999-2021) with CEA results were identified. 394 cases had histological confirmation as the diagnostic standard. Additionally, 237 cysts without histological confirmation demonstrated KRAS, GNAS, or RNF43 mutations by molecular testing and were combined with the histologically confirmed cysts for separate analysis on a total cohort of 631 cysts. RESULTS Median CEA was significantly higher in mucinous cysts (323.9 ng/mL, n=314) versus non-mucinous cysts (204.6 ng/mL, n=80) (p<0.001). Receiver operating characteristic curve analysis demonstrated an optimal CEA cut-off of 20 ng/mL (area under the curve: 80%), though the specificity was lower than desired (sensitivity 89%, specificity 64%). At the previously established threshold of 192 ng/mL, sensitivity and specificity were 56% and 78%, respectively. To achieve a specificity of 85% as originally reported, a CEA threshold of 250 ng/mL was needed; the 13 false positive cases at this threshold included 4 benign simple cysts, 2 squamoid cysts, 1 serous cystadenoma, 1 lymphoepithelial cyst and 5 more uncommon entities. All results remained similar within the total cohort after including additional cases with KRAS/GNAS/RNF43 mutations only. CONCLUSION Cyst fluid CEA continues to be a useful test in the diagnosis of mucinous pancreatic cysts but does not appear as specific as previously reported. Raising the CEA threshold to 250 ng/mL to maintain specificity for differentiating mucinous from non-mucinous cysts may be considered.
Collapse
Affiliation(s)
- Melanie C Kwan
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Martha Bishop Pitman
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Carlos Fernandez-Del Castillo
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - M Lisa Zhang
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
29
|
Ohtsuka T, Fernandez-Del Castillo C, Furukawa T, Hijioka S, Jang JY, Lennon AM, Miyasaka Y, Ohno E, Salvia R, Wolfgang CL, Wood LD. International evidence-based Kyoto guidelines for the management of intraductal papillary mucinous neoplasm of the pancreas. Pancreatology 2024; 24:255-270. [PMID: 38182527 DOI: 10.1016/j.pan.2023.12.009] [Citation(s) in RCA: 114] [Impact Index Per Article: 114.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/14/2023] [Accepted: 12/22/2023] [Indexed: 01/07/2024]
Abstract
This study group aimed to revise the 2017 international consensus guidelines for the management of intraductal papillary mucinous neoplasm (IPMN) of the pancreas, and mainly focused on five topics; the revision of high-risk stigmata (HRS) and worrisome features (WF), surveillance of non-resected IPMN, surveillance after resection of IPMN, revision of pathological aspects, and investigation of molecular markers in cyst fluid. A new development from the prior guidelines is that systematic reviews were performed for each one of these topics, and published separately to provide evidence-based recommendations. One of the highlights of these new "evidence-based guidelines" is to propose a new management algorithm, and one major revision is to include into the assessment of HRS and WF the imaging findings from endoscopic ultrasound (EUS) and the results of cytological analysis from EUS-guided fine needle aspiration technique, when this is performed. Another key element of the current guidelines is to clarify whether lifetime surveillance for small IPMNs is required, and recommends two options, "stop surveillance" or "continue surveillance for possible development of concomitant pancreatic ductal adenocarcinoma", for small unchanged BD-IPMN after 5 years surveillance. Several other points are also discussed, including identifying high-risk features for recurrence in patients who underwent resection of non-invasive IPMN with negative surgical margin, summaries of the recent observations in the pathology of IPMN. In addition, the emerging role of cyst fluid markers that can aid in distinguishing IPMN from other pancreatic cysts and identify those IPMNs that harbor high-grade dysplasia or invasive carcinoma is discussed.
Collapse
Affiliation(s)
- Takao Ohtsuka
- Department of Digestive Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.
| | | | - Toru Furukawa
- Department of Investigative Pathology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Susumu Hijioka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Jin-Young Jang
- Division of Hepatobiliary-Pancreatic Surgery, Departments of Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Anne Marie Lennon
- Division of Gastroenterology and Hepatology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Yoshihiro Miyasaka
- Department of Surgery, Fukuoka University Chikushi Hospital, and Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eizaburo Ohno
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
| | - Roberto Salvia
- Department of Surgery, Dentistry, Paediatrics and Gynaecology University of Verona, Verona, Italy
| | | | - Laura D Wood
- Departments of Pathology and Oncology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
30
|
Machicado JD, Napoleon B, Akshintala V, Bazarbashi AN, Bilal M, Corral JE, Dugum M, Han S, Hussain FS, Johnson AM, Jovani M, Kolb JM, Leonor P, Lee PJ, Mulki R, Shah H, Singh H, Sánchez-Luna SA, Shah SL, Singla A, Vargas EJ, Tielleman T, Nikahd M, Fry M, Culp S, Krishna SG. Structured training program on confocal laser endomicroscopy for pancreatic cystic lesions: a multicenter prospective study among early-career endosonographers (with video). Gastrointest Endosc 2023; 98:953-964. [PMID: 37473969 PMCID: PMC10771632 DOI: 10.1016/j.gie.2023.07.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/08/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND AND AIMS Data on how to teach endosonographers needle-based confocal laser endomicroscopy (nCLE)-guided histologic diagnosis of pancreatic cystic lesions (PCLs) are limited. Hence, we developed and tested a structured educational program to train early-career endosonographers in nCLE-guided diagnosis of PCLs. METHODS Twenty-one early-career nCLE-naïve endosonographers watched a teaching module outlining nCLE criteria for diagnosing PCLs. Participants then reviewed 80 high-yield nCLE videos, recorded diagnoses, and received expert feedback (phase 1). Observers were then randomized to a refresher feedback session or self-learning at 4 weeks. Eight weeks after training, participants independently assessed the same 80 nCLE videos without feedback and provided histologic predictions (phase 2). Diagnostic performance of nCLE to differentiate mucinous versus nonmucinous PCLs and to diagnose specific subtypes were analyzed using histopathology as the criterion standard. Learning curves were determined using cumulative sum analysis. RESULTS Accuracy and diagnostic confidence for differentiating mucinous versus nonmucinous PCLs improved as endosonographers progressed through nCLE videos in phase 1 (P < .001). Similar trends were observed with the diagnosis of PCL subtypes. Most participants achieved competency interpreting nCLE, requiring a median of 38 assessments (range, 9-67). During phase 2, participants independently differentiated PCLs with high accuracy (89%), high confidence (83%), and substantial interobserver agreement (κ = .63). Accuracy for nCLE-guided PCL subtype diagnoses ranged from 82% to 96%. The learned nCLE skills did not deteriorate at 8 weeks and were not impacted by a refresher session. CONCLUSIONS We developed a practical, effective, and durable educational intervention to train early-career endosonographers in nCLE-guided diagnosis of PCLs.
Collapse
Affiliation(s)
- Jorge D Machicado
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Bertrand Napoleon
- Gastroenterology Department, Hopital Privé Jean Mermoz, Ramsay Generale de Sante, Lyon, France
| | - Venkata Akshintala
- Division of Gastroenterology and Hepatology, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | | | - Mohammad Bilal
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, Minnesota, USA
| | - Juan E Corral
- Division of Gastroenterology and Hepatology, Presbyterian Hospital, Albuquerque, New Mexico, USA
| | | | - Samuel Han
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | | | - Alyson M Johnson
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA
| | - Manol Jovani
- Division of Gastroenterology, Maimonides Medical Center, SUNY Downstate University, Brooklyn, New York, USA
| | - Jennifer M Kolb
- Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Paul Leonor
- Division of Gastroenterology and Hepatology, Loma Linda University, Loma Linda, California, USA
| | - Peter J Lee
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Ramzi Mulki
- Basil I. Hirschowitz Endoscopic Center of Excellence, Division of Gastroenterology & Hepatology, Department of Internal Medicine, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA
| | - Hamza Shah
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Harkirat Singh
- Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Sergio A Sánchez-Luna
- Basil I. Hirschowitz Endoscopic Center of Excellence, Division of Gastroenterology & Hepatology, Department of Internal Medicine, The University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA
| | - Shawn L Shah
- Division of Digestive and Liver Diseases, Veterans Affairs North Texas Health Care System, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Anand Singla
- Division of Gastroenterology, University of Washington, Seattle, Washington, USA
| | - Eric J Vargas
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Thomas Tielleman
- Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Melica Nikahd
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Megan Fry
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Stacey Culp
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Somashekar G Krishna
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
31
|
Cui M, Hu Y, Zhang Z, Chen T, Dai M, Xu Q, Guo J, Zhang T, Liao Q, Yu J, Zhao Y. Cyst fluid glycoproteins accurately distinguishing malignancies of pancreatic cystic neoplasm. Signal Transduct Target Ther 2023; 8:406. [PMID: 37848412 PMCID: PMC10582020 DOI: 10.1038/s41392-023-01645-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 06/26/2023] [Accepted: 09/13/2023] [Indexed: 10/19/2023] Open
Abstract
Pancreatic cystic neoplasms (PCNs) are recognized as precursor lesions of pancreatic cancer, with a marked increase in prevalence. Early detection of malignant PCNs is crucial for improving prognosis; however, current diagnostic methods are insufficient for accurately identifying malignant PCNs. Here, we utilized mass spectrometry (MS)-based glycosite- and glycoform-specific glycoproteomics, combined with proteomics, to explore potential cyst fluid diagnostic biomarkers for PCN. The glycoproteomic and proteomic landscape of pancreatic cyst fluid samples from PCN patients was comprehensively investigated, and its characteristics during the malignant transformation of PCN were analyzed. Under the criteria of screening specific cyst fluid biomarkers for the diagnosis of PCN, a group of cyst fluid glycoprotein biomarkers was identified. Through parallel reaction monitoring (PRM)-based targeted glycoproteomic analysis, we validated these chosen glycoprotein biomarkers in a second cohort, ultimately confirming N-glycosylated PHKB (Asn-935, H5N2F0S0; Asn-935, H4N4F0S0; Asn-935, H5N4F0S0), CEACAM5 (Asn-197, H5N4F0S0) and ATP6V0A4 (Asn-367, H6N4F0S0) as promising diagnostic biomarkers for distinguishing malignant PCNs. These glycoprotein biomarkers exhibited robust performance, with an area under the curve ranging from 0.771 to 0.948. In conclusion, we successfully established and conducted MS-based glycoproteomic analysis to identify novel cyst fluid glycoprotein biomarkers for PCN. These findings hold significant clinical implications, providing valuable insights for PCN decision-making, and potentially offering therapeutic targets for PCN treatment.
Collapse
Affiliation(s)
- Ming Cui
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Disease, National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Ya Hu
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Disease, National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Zejian Zhang
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Disease, National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Tianqi Chen
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Disease, National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Menghua Dai
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Disease, National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Qiang Xu
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Disease, National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Junchao Guo
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Disease, National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Disease, National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Quan Liao
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Disease, National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Jun Yu
- Department of Medicine, Oncology, and Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
- Pancreas center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Yupei Zhao
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Disease, National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|