1
|
Dreyer SB, Beer P, Hingorani SR, Biankin AV. Improving outcomes of patients with pancreatic cancer. Nat Rev Clin Oncol 2025; 22:439-456. [PMID: 40329051 DOI: 10.1038/s41571-025-01019-9] [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: 04/04/2025] [Indexed: 05/08/2025]
Abstract
Research studies aimed at improving the outcomes of patients with pancreatic ductal adenocarcinoma (PDAC) have brought about limited progress, and in clinical practice, the optimized use of surgery, chemotherapy and supportive care have led to modest improvements in survival that have probably reached a plateau. As a result, PDAC is expected to be the second leading cause of cancer-related death in Western societies within a decade. The development of therapeutic advances in PDAC has been challenging owing to a lack of actionable molecular targets, a typically immunosuppressive microenvironment, and a disease course characterized by rapid progression and clinical deterioration. Yet, the progress in our understanding of PDAC and identification of novel therapeutic opportunities over the past few years is leading to a strong sense of optimism in the field. In this Perspective, we address the aforementioned challenges, including biological aspects of PDAC that make this malignancy particularly difficult to treat. We explore specific areas with potential for therapeutic advances, including targeting mutant KRAS, novel strategies to harness the antitumour immune response and approaches to early detection, and propose mechanisms to improve clinical trial design and to overcome various community and institutional barriers to progress.
Collapse
Affiliation(s)
- Stephan B Dreyer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- Department of Hepatobiliary Surgery, Royal Liverpool University Hospital, Liverpool, UK
| | - Philip Beer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- Hull York Medical School, University of York, York, UK
| | - Sunil R Hingorani
- Department of Internal Medicine, Division of Hemotology/Oncology, University of Nebraska Medical Center, Omaha, NE, USA
- Pancreatic Cancer Center of Excellence, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK.
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK.
| |
Collapse
|
2
|
Bilreiro C, Fernandes FF, Simões RV, Henriques R, Chavarrías C, Ianus A, Castillo-Martin M, Carvalho T, Matos C, Shemesh N. Pancreatic Intraepithelial Neoplasia Revealed by Diffusion-Tensor MRI. Invest Radiol 2025; 60:397-406. [PMID: 39668406 DOI: 10.1097/rli.0000000000001142] [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: 12/14/2024]
Abstract
OBJECTIVES Detecting premalignant lesions for pancreatic ductal adenocarcinoma, mainly pancreatic intraepithelial neoplasia (PanIN), is critical for early diagnosis and for understanding PanIN biology. Based on PanIN's histology, we hypothesized that diffusion tensor imaging (DTI) and T2* could detect PanIN. MATERIALS AND METHODS DTI was explored for the detection and characterization of PanIN in genetically engineered mice (KC, KPC). Following in vivo DTI, ex vivo ultrahigh-field (16.4 T) MR microscopy using DTI, T2* was performed with histological validation. Sources of MR contrasts and histological features were investigated, including histological scoring for disease burden (lesion span) and severity (adjusted score). To test if findings in mice can be translated to humans, human pancreas specimens were imaged. RESULTS DTI detected PanIN and pancreatic ductal adenocarcinoma in vivo (6 KPC, 4 KC, 6 controls) with high discriminative ability: fractional anisotropy (FA) and radial diffusivity with area under the curve = 0.983 (95% confidence interval: 0.932-1.000); mean diffusivity and axial diffusivity (AD) with area under the curve = 1 (95% confidence interval: 1.000-1.000). MR microscopy with histological correlation (20 KC/KPC; 5 controls) revealed that sources of MR contrasts likely arise from microarchitectural signatures: high FA, AD in fibrotic areas surrounding lesions, high diffusivities within cysts, and high T2* within lesions' stroma. The strongest histological correlations for lesion span and adjusted score were obtained with AD ( R = 0.708, P < 0.001; R = 0.789, P < 0.001, respectively). Ex vivo observations in 5 human pancreases matched our findings in mice, revealing substantial contrast between PanIN and normal pancreas. CONCLUSIONS DTI and T2* are useful for detecting and characterizing PanIN in genetically engineered mice and in the human pancreas, especially with AD and FA. These are encouraging findings for future clinical applications of pancreatic imaging.
Collapse
Affiliation(s)
- Carlos Bilreiro
- From the Radiology Department, Champalimaud Foundation, Lisbon, Portugal (C.B., C.M.); Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal (C.B., F.F.F., R.H., C.C., A.I., M.C.-M., T.C., C.M., N.S.); Nova Medical School, Lisbon, Portugal (C.B.); i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal (R.V.S.); and Pathology Department, Champalimaud Foundation, Lisbon, Portugal (M.C.-M.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Hruban C, Bruhm DC, Chen IM, Koul S, Annapragada AV, Vulpescu NA, Short S, Theile S, Boyapati K, Alipanahi B, Skidmore ZL, Leal A, Cristiano S, Adleff V, Johannsen JS, Scharpf RB, Foda ZH, Phallen J, Velculescu VE. Genome-wide analyses of cell-free DNA for therapeutic monitoring of patients with pancreatic cancer. SCIENCE ADVANCES 2025; 11:eads5002. [PMID: 40397745 DOI: 10.1126/sciadv.ads5002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 04/07/2025] [Indexed: 05/23/2025]
Abstract
Determining response to therapy for patients with pancreatic cancer can be challenging. We evaluated methods for assessing therapeutic response using cell-free DNA (cfDNA) in plasma from patients with metastatic pancreatic cancer in the CheckPAC trial (NCT02866383). Patients were evaluated before and after initiation of therapy using tumor-informed plasma whole-genome sequencing (WGMAF) and tumor-independent genome-wide cfDNA fragmentation profiles and repeat landscapes (ARTEMIS-DELFI). Using WGMAF, molecular responders had a median overall survival (OS) of 319 days compared to 126 days for nonresponders [hazard ratio (HR) = 0.29, 95% confidence interval (CI) = 0.11-0.79, P = 0.011]. For ARTEMIS-DELFI, patients with low scores after therapy initiation had longer median OS than patients with high scores (233 versus 172 days, HR = 0.12, 95% CI = 0.046-0.31, P < 0.0001). We validated ARTEMIS-DELFI in patients with pancreatic cancer in the PACTO trial (NCT02767557). These analyses suggest that noninvasive mutation and fragmentation-based cfDNA approaches can identify therapeutic response of individuals with pancreatic cancer.
Collapse
Affiliation(s)
- Carolyn Hruban
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel C Bruhm
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Inna M Chen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Shashikant Koul
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akshaya V Annapragada
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas A Vulpescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah Short
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susann Theile
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Kavya Boyapati
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | - Alessandro Leal
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephen Cristiano
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vilmos Adleff
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julia S Johannsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Medicine, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Robert B Scharpf
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachariah H Foda
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Victor E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Mukherjee S, Antony A, Patnam NG, Trivedi KH, Karbhari A, Nagaraj M, Murlidhar M, Goenka AH. Pancreas segmentation using AI developed on the largest CT dataset with multi-institutional validation and implications for early cancer detection. Sci Rep 2025; 15:17096. [PMID: 40379726 PMCID: PMC12084540 DOI: 10.1038/s41598-025-01802-9] [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] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 05/08/2025] [Indexed: 05/19/2025] Open
Abstract
Accurate and fully automated pancreas segmentation is critical for advancing imaging biomarkers in early pancreatic cancer detection and for biomarker discovery in endocrine and exocrine pancreatic diseases. We developed and evaluated a deep learning (DL)-based convolutional neural network (CNN) for automated pancreas segmentation using the largest single-institution dataset to date (n = 3031 CTs). Ground truth segmentations were performed by radiologists, which were used to train a 3D nnU-Net model through five-fold cross-validation, generating an ensemble of top-performing models. To assess generalizability, the model was externally validated on the multi-institutional AbdomenCT-1K dataset (n = 585), for which volumetric segmentations were newly generated by expert radiologists and will be made publicly available. In the test subset (n = 452), the CNN achieved a mean Dice Similarity Coefficient (DSC) of 0.94 (SD 0.05), demonstrating high spatial concordance with radiologist-annotated volumes (Concordance Correlation Coefficient [CCC]: 0.95). On the AbdomenCT-1K dataset, the model achieved a DSC of 0.96 (SD 0.04) and a CCC of 0.98, confirming its robustness across diverse imaging conditions. The proposed DL model establishes new performance benchmarks for fully automated pancreas segmentation, offering a scalable and generalizable solution for large-scale imaging biomarker research and clinical translation.
Collapse
Affiliation(s)
- Sovanlal Mukherjee
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ajith Antony
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Nandakumar G Patnam
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Kamaxi H Trivedi
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Aashna Karbhari
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Madhu Nagaraj
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Murlidhar Murlidhar
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Ajit H Goenka
- Professor of Radiology, Consultant, Divisions of Abdominal and Nuclear Radiology, Co-Chair, Nuclear Radiology Research Operations, Chair, Enterprise PET/MR Research, Education and Executive Committee, Program Co-Leader, Risk Assessment, Early Detection and Interception (REDI), Mayo Clinic Comprehensive Cancer Center (MCCCC), 200 First St SW, Charlton 1, Rochester, MN, 55905, USA.
| |
Collapse
|
5
|
Lee AA, Twoy A, Sutton A, Fushimi N, Park WG. Suboptimal adherence to surveillance in high-risk individuals for pancreatic cancer at a tertiary care academic center: Lessons from real-world surveillance patterns. Pancreatology 2025:S1424-3903(25)00087-0. [PMID: 40382255 DOI: 10.1016/j.pan.2025.05.002] [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: 03/25/2025] [Revised: 05/02/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND/OBJECTIVES Screening for pancreatic ductal adenocarcinoma alters the course of disease among high-risk individuals (HRIs) and is recommended by multiple societies including the International Cancer of the Pancreas Screening Consortium, American Gastroenterological Association, and American Society of Gastrointestinal Endoscopy. However, there are limited analyses on the screening rates and barriers to adherence among HRIs. This study aims to describe real-world longitudinal screening adherence of a HRI surveillance cohort and identify potential barriers to adherence. METHODS Patients followed by Stanford's Benign Pancreas Clinic were identified as HRIs if they met screening criteria per the latest abovementioned screening guidelines, and were included in our study if they underwent at least 1 screening exam. Data on HRIs were obtained retrospectively from our hospital's electronic health record system. Patient and screening characteristics were analyzed by adherence rates. RESULTS 262 HRIs undergoing recommended screening were identified. The mean follow-up time per patient was 4.9 years and the mean successful annual screening rate within the entire cohort was 67%. HRIs in the lowest quartile of adherence were more likely to have more EUS rather than MRI surveillance exams compared to those who were completely adherent (p = 0.01). HRIs who were completely adherent were also an older cohort compared to those with non-complete adherence (p = 0.02) or in the lowest quartile of adherence (p = 0.01). CONCLUSIONS It is difficult to achieve high adherence rates for annual pancreatic cancer screening of HRIs as recommended by the latest guidelines. Age and screening modality may be factors that contribute to adherence difficulty.
Collapse
Affiliation(s)
- Alice A Lee
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA.
| | - Abigail Twoy
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Audrey Sutton
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Naoko Fushimi
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Walter G Park
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| |
Collapse
|
6
|
Stoop TF, Javed AA, Oba A, Koerkamp BG, Seufferlein T, Wilmink JW, Besselink MG. Pancreatic cancer. Lancet 2025; 405:1182-1202. [PMID: 40187844 DOI: 10.1016/s0140-6736(25)00261-2] [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: 10/02/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 04/07/2025]
Abstract
Pancreatic cancer is frequently a lethal disease with an aggressive tumour biology often presenting with non-specific symptoms. Median survival is approximately 4 months with a 5-year survival of 13%. Surveillance is recommended in individuals with familial pancreatic cancer, specific mutations, and high-risk intraductal papillary mucinous neoplasm, as they are at high risk of developing pancreatic cancer. Chemotherapy combined with surgical resection remains the cornerstone of treatment. However, only a small subset of patients are candidates for surgery. Multi-agent chemotherapy has improved survival in the palliative setting for patients with metastatic disease, as (neo)adjuvant and induction therapy have in patients with borderline resectable and locally advanced pancreatic. Given that pancreatic cancer is predicted to become the second leading cause of cancer-related death by 2030, novel therapies are urgently needed.
Collapse
Affiliation(s)
- Thomas F Stoop
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Ammar A Javed
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands; Division of Surgical Oncology, Department of Surgery, New York University Medical Center, New York, NY, USA
| | - Atsushi Oba
- Department of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake, Tokyo, Japan; Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan; Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Thomas Seufferlein
- Department of International Medicine I, Ulm University Hospital, Ulm, Germany
| | - Johanna W Wilmink
- Department of Medical Oncology, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Marc G Besselink
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
7
|
Bilreiro C, Andrade L, Henriques R, Loução N, Matos C, Shemesh N. Diffusion tensor imaging and diffusion kurtosis imaging of the pancreas - feasibility, robustness and protocol comparison in a healthy population. Abdom Radiol (NY) 2025:10.1007/s00261-025-04889-w. [PMID: 40137946 DOI: 10.1007/s00261-025-04889-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025]
Abstract
PURPOSE This study aims to determine the feasibility, image quality, intra-subject repeatability and inter-reader variability of Diffusion tensor (DTI) and Diffusion kurtosis imaging (DKI) for pancreatic imaging using different protocols and report normative values in healthy individuals. METHODS Single-institution prospective study performed on healthy volunteers in a clinical 3T scanner, using two different protocols (6/16 diffusion directions). Acquisitions were repeated twice to assess intra-subject repeatability. To assess inter-reader variability, Mean diffusivity (MD), Axial diffusivity (AD), Radial diffusivity (RD), Apparent diffusion coefficient (ADC) and Mean kurtosis (MK) values were extracted from segmented pancreas by two radiologists. A Likert scale was used by both readers to assess subjective image quality. RESULTS Twelve healthy volunteers were recruited for each MRI protocol. The 6 diffusion directions protocol was shorter: 7 min vs. 14 min (corresponding to 4 min vs. 7.5 min for a DTI only reconstruction). No differences in image quality were found between protocols. Only MK maps showed implausible estimates, leading to the exclusion of median 16% and 17.7% pixels for the 6- and 16-direction protocols, respectively. Intra-subject repeatability was determined with negligible coefficients of repeatability for DTI; however, MK presented slightly higher values. Inter-reader agreement was excellent for all maps (ICC > 0.9). CONCLUSIONS DTI and DKI of the pancreas are feasible in clinical settings, with excellent inter-observer agreement and good image quality. Intra-subject repeatability is excellent for DTI, but some variability was observed with DKI. A 6-directions protocol may be preferred due to faster acquisition without quantitatively compromising estimates. MK inaccuracies prompt further research for improving artifact correction.
Collapse
Affiliation(s)
- Carlos Bilreiro
- Champalimaud Foundation, Lisbon, Portugal.
- Universidade Nova de Lisboa, Lisbon, Portugal.
| | | | | | | | | | | |
Collapse
|
8
|
Song W, Yu Y, Wang S, Cui Z, Zhu Q, Liu W, Wei S, Chi J. Metabolic reprogramming shapes the immune microenvironment in pancreatic adenocarcinoma: prognostic implications and therapeutic targets. Front Immunol 2025; 16:1555287. [PMID: 40191200 PMCID: PMC11968369 DOI: 10.3389/fimmu.2025.1555287] [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] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 02/17/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Pancreatic adenocarcinoma (PAAD) is characterized by a profoundly immunosuppressive tumor microenvironment (TME) that limits the efficacy of immunotherapy. Emerging evidence suggests that tumor-specific metabolic reprogramming may drive disease progression and shape the immune landscape in PAAD. Methods We integrated multi-omics data from TCGA, GEO, and ICGC to identify key metabolism-related genes (MRGs) that influence immune cell infiltration, tumor progression, and patient survival. Based on nine pivotal MRGs (including ANLN, PKMYT1, and HMGA1), we developed and validated a novel metabolic-prognostic index (MPI). Functional enrichment analyses were conducted to elucidate the metabolic pathways associated with different MPI risk groups. In vitro experiments and drug sensitivity analyses were performed to confirm the oncogenic role of selected MRGs and to explore their therapeutic implications. Results The MPI effectively stratified patients into high- and low-risk groups. High-MPI scores correlated with poor overall survival, elevated tumor mutation burden (TMB), and an immunosuppressive TME, evidenced by reduced CD8⁺ T-cell infiltration and increased expression of immune checkpoints (PD-L1, TGF-β). Functional enrichment revealed glycolysis and folate biosynthesis as dominant pathways in high-MPI groups, whereas fatty acid metabolism prevailed in low-MPI groups. Experimental validation underscored the role of ANLN in promoting epithelial-mesenchymal transition (EMT) and immune evasion via NF-κB signaling. ANLN knockdown significantly reduced glycolytic activity, tumor cell migration, and immune evasion. Drug sensitivity analyses indicated resistance to gemcitabine but sensitivity to afatinib in high-MPI patients. Although TIDE analysis predicted immune checkpoint inhibitor (ICI) resistance in high-MPI tumors, a subset of patients showed favorable responses to anti-PD-L1 therapy. Discussion These findings provide a comprehensive framework for understanding how metabolic reprogramming shapes PAAD's immunosuppressive TME and affects treatment outcomes. By accurately stratifying patients, the MPI serves as a promising tool to guide therapeutic decisions, including targeted therapy selection and immunotherapy prediction, ultimately offering potential for more personalized management of PAAD.
Collapse
Affiliation(s)
- Weihua Song
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yabin Yu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Huai’an No. 1 People’s Hospital of Nanjing Medical University, Huai’an, Jiangsu, China
| | - Siqi Wang
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhengyi Cui
- Department of Public Health, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Qiusi Zhu
- College of Animal Science and Technology, Northeast Agricultural University, Haerbin, China
| | - Wangrui Liu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shiyin Wei
- Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Key Laboratory of Tumor Molecular Pathology of Baise, Baise, China
| | - Jiachang Chi
- Department of Thoracic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
9
|
Mohamed G, Munir M, Rai A, Gaddam S. Pancreatic Cancer: Screening and Early Detection. Gastroenterol Clin North Am 2025; 54:205-221. [PMID: 39880528 DOI: 10.1016/j.gtc.2024.09.006] [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 cancer, often diagnosed at advanced stages, has poor survival rates. Effective screening aims to detect the disease early, improving outcomes. Current guidelines recommend screening high-risk groups, including those with a family history or genetic predispositions, using methods like endoscopic ultrasound and MRI. The American Gastroenterological Association and other organizations advise annual surveillance for high-risk individuals, typically starting at the age of 50 or 10 years younger than the youngest affected relative. For certain genetic syndromes, such as Peutz-Jeghers syndrome or hereditary pancreatitis, screening may begin as early as the age of 35 to 40 years.
Collapse
Affiliation(s)
- Ghada Mohamed
- Department of Internal Medicine, Lahey Hospital & Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Malak Munir
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, ST, Suite 7705, Los Angeles, CA 90048, USA
| | - Amar Rai
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, ST, Suite 7705, Los Angeles, CA 90048, USA
| | - Srinivas Gaddam
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, ST, Suite 7705, Los Angeles, CA 90048, USA.
| |
Collapse
|
10
|
Heller M, Mann DA, Katona BW. Current Approaches of Pancreatic Cancer Surveillance in High-Risk Individuals. J Gastrointest Cancer 2025; 56:61. [PMID: 39932614 PMCID: PMC11814005 DOI: 10.1007/s12029-025-01184-1] [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] [Accepted: 01/25/2025] [Indexed: 02/14/2025]
Abstract
Currently, those recommended to undergo pancreatic cancer (PC) surveillance include appropriately aged individuals at high risk of PC due to an identifiable genetic susceptibility or those without identifiable genetic susceptibility who nonetheless have a strong family history of PC. With increases in identification of individuals at high risk for PC and increased use of PC surveillance in clinical practice, there has been increasing debate about who should undergo surveillance as well as how surveillance should be performed including use of imaging and blood-based testing. Furthermore, there is increasing interest in the outcomes of PC surveillance in high-risk individuals with some studies demonstrating that surveillance leads to downstaging of PC and improvements in survival. In this review, we summarize the current state of PC surveillance in high-risk individuals, providing an overview of the risk factors associated with PC, selection of high-risk individuals for PC surveillance, and the current, but non-uniform, recommendations for performing PC surveillance. Additionally, we review approaches to apply various imaging and blood-based tests to surveillance and the outcomes of PC surveillance.
Collapse
Affiliation(s)
- Melissa Heller
- Division of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Derek A Mann
- Division of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bryson W Katona
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd., 751 South Pavilion, Philadelphia, PA, 19104, USA.
| |
Collapse
|
11
|
Bogdanski AM, Acedo P, Wallace MB, van Leerdam ME, Klatte DCF. Recommendations, evidence and sustainability of screening for pancreatic cancer in high-risk individuals. Best Pract Res Clin Gastroenterol 2025; 74:101974. [PMID: 40210328 DOI: 10.1016/j.bpg.2025.101974] [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: 12/10/2024] [Accepted: 12/31/2024] [Indexed: 04/12/2025]
Abstract
Pancreatic cancer is a highly lethal malignancy and is predicted to become the second leading cause of cancer-related deaths by 2030. Early detection significantly improves outcomes, but general population screening remains infeasible due to the low prevalence of the disease and lack of specific biomarkers. This review evaluates current recommendations for pancreatic cancer surveillance in high-risk individuals, synthesises evidence from recent studies and explores the sustainability of current imaging-based surveillance programmes. Challenges such as overdiagnosis, economic feasibility and disparities in access highlight the need for targeted, cost-effective strategies. Collaborative initiatives and consortia are needed to advance biomarker research and refine risk stratification. By integrating evidence-based recommendations with sustainable approaches, this review outlines pathways to improve early detection and reduce mortality from pancreatic cancer.
Collapse
Affiliation(s)
- Aleksander M Bogdanski
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, United States; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Pilar Acedo
- Institute for Liver and Digestive Health, Division of Medicine, University College London, London, United Kingdom
| | - Michael B Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, United States
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands; Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Derk C F Klatte
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, United States; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
12
|
Hussein T, Mátrai P, Vass V, Szentesi A, Hegyi P. Onset of pancreatic cancer before and after acute pancreatitis: A multicenter longitudinal cohort study. Pancreatology 2025; 25:29-34. [PMID: 39734119 DOI: 10.1016/j.pan.2024.12.007] [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: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Pancreatic cancer (PC) is a leading cause of cancer mortality, often diagnosed at advanced stages. Acute pancreatitis (AP), particularly idiopathic cases, may serve as an early indicator of PC. OBJECTIVE This multicenter cohort study investigated the incidence of PC before and after an AP episode, focusing on idiopathic AP and the role of pseudocysts as potential early markers for PC development. METHODS We analyzed data from 2356 AP patients across 25 centers, with a median follow-up of 4.1 years (IQR: 1.6-6.8 years). Patients were categorized into 'PC before AP' and 'PC after AP' groups, and relative risk (RR) and adjusted odds ratios (OR) were calculated for idiopathic AP cases to quantify PC risk. RESULTS Among all cases, 69 patients (2.9 %) developed PC: 1.4 % (n = 34) before and 1.5 % (n = 35) after AP. Idiopathic AP cases had a fourfold higher risk of PC (OR = 4.46, [2.25-8.85]). Notably, pseudocysts were five times more prevalent in the PC group (14 %) compared to controls (3 %) (RR = 5.66; p < 0.01), often located at the tumor site. PC developed in 3 % of idiopathic AP cases versus 1.0 % in non-idiopathic cases. The median time to PC diagnosis post-AP was 373 days. CONCLUSION Idiopathic AP and pseudocyst formation significantly elevate the risk of PC, particularly within two years. These findings underscore the need for structured follow-up and early screening in idiopathic AP cases to improve PC detection and survival outcomes.
Collapse
Affiliation(s)
- Tamás Hussein
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary; Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Mátrai
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Vivien Vass
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Andrea Szentesi
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Hegyi
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary; Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary; Centre for Translational Medicine, Semmelweis University, Budapest, Hungary; Translational Pancreatology Research Group, Interdisciplinary Centre of Excellence for Research Development and Innovation, University of Szeged, Szeged, Hungary.
| |
Collapse
|
13
|
Katona BW, Lubinski J, Pal T, Huzarski T, Foulkes WD, Moller P, Eisen A, Randall Armel S, Neuhausen SL, Raj R, Aeilts A, Singer CF, Bordeleau L, Karlan B, Olopade O, Tung N, Zakalik D, Kotsopoulos J, Fruscio R, Eng C, Sun P, Narod SA. The incidence of pancreatic cancer in women with a BRCA1 or BRCA2 mutation. Cancer 2025; 131:e35666. [PMID: 39611336 DOI: 10.1002/cncr.35666] [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: 05/06/2024] [Revised: 07/17/2024] [Accepted: 08/12/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND The lifetime risk of pancreatic cancer in women with a germline mutation in BRCA1 and BRCA2 is not well established. In an international prospective cohort of female carriers of BRCA1 and BRCA2 mutations, the cumulative incidence of pancreatic cancer from age 40 until 80 years was estimated. METHODS A total of 8295 women with a BRCA1 or BRCA2 mutation were followed for new cases of pancreatic cancer. Subjects were followed from the date of baseline questionnaire or age 40 years (whichever came last) until a new diagnosis of pancreatic cancer, death from another cause, or date of last follow-up. RESULTS Thirty-four incident pancreatic cancer cases were identified in the cohort. The annual risk of pancreatic cancer between age 40 and 80 years was 0.04% for BRCA1 carriers and 0.09% for BRCA2 carriers. Via the Kaplan-Meier method, the cumulative incidence from age 40 to 80 years was 2.2% (95% CI, 1.1%-4.3%) for BRCA1 carriers and 2.7% (95% CI, 1.3%-5.4%) for BRCA2 carriers. Only two of the 34 cases reported a first-degree relative with pancreatic cancer (hazard ratio, 4.75; 95% CI, 1.13-19.9; p = .03). Risk factors for pancreatic cancer included alcohol intake and a history of diabetes. The 5-year survival rate for the 34 cases was 8.8%. CONCLUSIONS The lifetime risk of pancreatic cancer is approximately 2% in women with a BRCA1 mutation and 3% for women with a BRCA2 mutation. The poor survival in hereditary pancreatic cancer underscores the need for novel antitumoral strategies.
Collapse
Affiliation(s)
- Bryson W Katona
- Basser Center for BRCA and Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jan Lubinski
- International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Tuya Pal
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tomasz Huzarski
- International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - William D Foulkes
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Pal Moller
- Institute of Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Andrea Eisen
- Sunnybrook Regional Cancer Centre, Toronto, Ontario, Canada
| | - Susan Randall Armel
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
- Division of Gynecologic Oncology, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Susan L Neuhausen
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - Rebecca Raj
- Women's College Research Institute, Toronto, Ontario, Canada
| | - Amber Aeilts
- Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, Ohio, USA
| | - Christian F Singer
- Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Louise Bordeleau
- Department of Oncology, Juravinski Cancer Centre, Hamilton, Ontario, Canada
| | - Beth Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Olufunmilayo Olopade
- Department of Medicine and Human Genetics, University of Chicago, Chicago, Illinois, USA
| | - Nadine Tung
- Cancer Risk and Prevention Program, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Dana Zakalik
- Cancer Genetics Program, Beaumont Hospital, Royal Oak, Michigan, USA
| | - Joanne Kotsopoulos
- Women's College Research Institute, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Robert Fruscio
- Department of Medicine and Surgery, University of Milan Bicocca, IRCCS San Gerardo, Monza, Italy
| | - Charis Eng
- Center for Personalized Genetic Healthcare, Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ping Sun
- Women's College Research Institute, Toronto, Ontario, Canada
| | - Steven A Narod
- Women's College Research Institute, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Du Q, Zhang M, Gao A, He T, Guo M. Epigenetic silencing ZSCAN23 promotes pancreatic cancer growth by activating Wnt signaling. Cancer Biol Ther 2024; 25:2302924. [PMID: 38226836 PMCID: PMC10793710 DOI: 10.1080/15384047.2024.2302924] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/04/2024] [Indexed: 01/17/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most malignant tumor. Zinc finger and SCAN domain-containing protein 23 (ZSCAN23) is a new member of the SCAN domain family. The expression regulation and biological function remain to be elucidated. In this study, we explored the epigenetic regulation and the function of ZSCAN23 in PDAC. ZSCAN23 was methylated in 60.21% (171/284) of PDAC and its expression was regulated by promoter region methylation. The expression of ZSCAN23 inhibited cell proliferation, colony formation, migration, invasion, and induced apoptosis and G1/S phase arrest. ZSCAN23 suppressed Panc10.05 cell xenograft growth in mice. Mechanistically, ZSCAN23 inhibited Wnt signaling by interacting with myosin heavy chain 9 (MYH9) in pancreatic cancer cells. ZSCAN23 is frequently methylated in PDAC and may serve as a detective marker. ZSCAN23 suppresses PDAC cell growth both in vitro and in vivo.
Collapse
Affiliation(s)
- Qian Du
- Department of Gastroenterology and Hepatology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Meiying Zhang
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Aiai Gao
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Tao He
- Department of Pathology, Characteristic Medical Center of the Chinese People's Armed Police Force, Tianjin, People's Republic of China
| | - Mingzhou Guo
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- National Key Laboratory of Kidney Diseases, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
15
|
Gros L, Yip R, Zhu Y, Li P, Paksashvili N, Sun Q, Yankelevitz DF, Henschke CI. GI cancer mortality in participants in low dose CT screening for lung cancer with a focus on pancreatic cancer. Sci Rep 2024; 14:29851. [PMID: 39617764 PMCID: PMC11609297 DOI: 10.1038/s41598-024-76322-z] [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] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/14/2024] [Indexed: 12/06/2024] Open
Abstract
Lung cancer, the leading cause of cancer deaths globally, has better survival rates with early detection. Annual low-dose CT (LDCT) screenings are recommended for high-risk individuals due to age and smoking. These individuals are also at risk for other cancers. Our study explores gastrointestinal (GI) cancer mortality in lung cancer screening participants and the potential of LDCT screenings to detect pancreatic cancer. We utilized data from a prospective multi-institutional cohort study, the International Early Lung Cancer Action Project (I-ELCAP). We analyzed GI cancer deaths among participants in New York State (1992-2010), exploring demographics and GI cancer distribution. Radiologists retrospectively reviewed pancreatic cancer cases within 24 months post-LDCT, comparing findings with original reports. Among 10,150 participants, 189 died from GI cancers; mean age 75, mostly male smokers. Pancreatic cancer (41.8%) led, followed by esophageal (17.5%) and colon cancer (16.9%). Median time between baseline LDCT and death was 116 months (9.7 years). 82/189 (43.4%) participants died within 5 years of their last LDCT screening, with pancreatic cancer again prominent (45.1%). In 79 pancreatic cancer deaths, 17.7% occurred within 24 months post-LDCT. A re-review identified previously undetected pancreatic findings, with 4 out of 14 participants (28.6%) showing abnormalities. This underscores the potential of lung cancer screening programs to provide insights beyond lung health. This study of over 10,000 participants in a lung cancer screening program reveals that they are at risk for GI cancer deaths, particularly pancreatic cancer. Re-reviews of LDCT scans revealed previously undocumented pancreatic findings in a third of participants who died from pancreatic cancer, underscoring the need to identify, document, and follow up on these findings.
Collapse
Affiliation(s)
- Louis Gros
- Department of Radiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Rowena Yip
- Department of Radiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Yeqing Zhu
- Department of Radiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Pengfei Li
- Department of Radiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Natela Paksashvili
- Department of Radiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Qi Sun
- Department of Radiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - David F Yankelevitz
- Department of Radiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Claudia I Henschke
- Department of Radiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
| |
Collapse
|
16
|
Beck SH, Karstensen JG, Bülow S, Andersen KK, Hansen TVO, Højen H, Jespersen N, Kuhlmann TP, Pommergaard HC, Wewer MD, Wullum L, Jelsig AM, Burisch J. Cancer Risks in Attenuated and Classical Familial Adenomatous Polyposis: A Nationwide Cohort With Matched, Nonexposed Individuals. Am J Gastroenterol 2024:00000434-990000000-01415. [PMID: 39471488 DOI: 10.14309/ajg.0000000000003167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 10/25/2024] [Indexed: 11/01/2024]
Abstract
INTRODUCTION Familial adenomatous polyposis (FAP) is caused by pathogenic variants in the APC gene. FAP is usually categorized according to phenotype: classical FAP (CFAP) and attenuated FAP (AFAP); the latter is considered to have a milder disease course. We aimed to assess the risk of overall and specific cancers in patients with CFAP and AFAP compared with matched, nonexposed individuals. METHODS All known Danish patients with FAP were classified as either CFAP or AFAP and assigned 4 matched, nonexposed individuals. The risk of overall and specific cancers, and mortality were analyzed. RESULTS The analysis included 311 patients with CFAP, 134 patients with AFAP, and 1,600 nonexposed individuals. The overall cancer risk was significantly higher for both patients with CFAP and AFAP than for nonexposed individuals, with hazard ratios (HRs) of 4.77 (95% confidence interval [CI], 3.61-6.32; P < 0.001) for CFAP and 3.22 (95% CI, 2.16-4.80; P < 0.001) for AFAP. No significant difference was observed when comparing CFAP and AFAP (HR = 1.48; 95% CI, 0.98-2.25; P = 0.0646). The HR of colonic cancer was 2.16 (95% CI, 0.99-7.72; P = 0.0522) and 2.72 (95% CI, 1.19-6.22; P = 0.0177 for CFAP and AFAP), respectively, compared with nonexposed and did not differ between patients with CFAP and AFAP (HR = 0.80; 95% CI, 0.32-2.00; P = 0.6278). Mortality was significantly higher in CFAP (HR = 2.96; 95% CI, 2.04-4.28; P < 0.001), but not in AFAP (HR = 1.40; 95% CI, 0.73-2.69; P = 0.311). DISCUSSION Nationwide data reveal differing risk profiles for specific cancers and mortality in AFAP and CFAP compared with nonexposed individuals. The cancer burden of AFAP necessitates consistent monitoring of these patients.
Collapse
Affiliation(s)
- Søren Hammershøj Beck
- Danish Polyposis Register, Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | - John Gásdal Karstensen
- Danish Polyposis Register, Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Steffen Bülow
- Danish Polyposis Register, Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | | | - Thomas van Overeem Hansen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Genetics, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Helle Højen
- Danish Polyposis Register, Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | - Niels Jespersen
- Danish Polyposis Register, Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | - Tine Plato Kuhlmann
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Pathology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Hans Christian Pommergaard
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Surgery and Transplantation, Copenhagen University Hospital-Rigshospitalet, Hepatic Malignancy Surgical Research Unit (HEPSURU), Copenhagen, Denmark
| | - Mads Damsgaard Wewer
- Danish Polyposis Register, Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- Gastro Unit, Medical Division, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | | | - Anne Marie Jelsig
- Department of Clinical Genetics, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Johan Burisch
- Danish Polyposis Register, Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Gastro Unit, Medical Division, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| |
Collapse
|
17
|
Wang Q, Liang T, Yang W, Xu Y, Qin C, Han H, Zhou X, Wang Y, Wang Z, Hu N. A smart tablet-phone-based high-performance pancreatic cancer cell biosensing system for drug screening. Talanta 2024; 278:126484. [PMID: 38941810 DOI: 10.1016/j.talanta.2024.126484] [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: 05/22/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Exploring more efficient pancreatic cancer drug screening platforms is of significant importance for accelerating the drug development process. In this study, we developed a high-sensitivity bioluminescence system based on smartphones and smart tablets, and constructed a pancreatic cancer drug screening platform (PCDSP) by combining the pancreatic cancer cell sensing model (PCCSM) on the multiwell plates (MTP). A smart tablet was used as the light source and a smartphone as the colorimetric sensing device. The smartphone dynamically controls the color and brightness displayed on the smart tablet to achieve lower LOD and wider detection ranges. We constructed PCCSM for 24 h, 48 h, and 72 h , and performed colorimetric experiments using both PCDSP and a commercial plate reader (CPR). The results showed that the PCDSP had a lower LOD than that of CPR. Moreover, PCDSP even exhibited a lower LOD for 24 h PCCSM testing compared to CPR for 48 h PCCSM testing, effectively shortening the drug evaluation process. Additionally, the PCDSP offers higher portability and efficiency compared with CPR, making it a promising platform for efficient pancreatic cancer drug screening.
Collapse
Affiliation(s)
- Qiang Wang
- Department of General Surgery, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang Provincial People's Hospital), Hangzhou Medical College, Taizhou, 317200, Zhejiang, China
| | - Tao Liang
- Laboratory Medicine Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China; Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, China
| | - Wenjian Yang
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, 310024, China
| | - Youjian Xu
- Department of General Surgery, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang Provincial People's Hospital), Hangzhou Medical College, Taizhou, 317200, Zhejiang, China
| | - Chunlian Qin
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, China
| | - Haote Han
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, China.
| | - Xiyang Zhou
- Department of General Surgery, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang Provincial People's Hospital), Hangzhou Medical College, Taizhou, 317200, Zhejiang, China.
| | - Yingwei Wang
- Department of Laboratory Medicine, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang Provincial People's Hospital), Hangzhou Medical College, Taizhou, 317200, Zhejiang, China.
| | - Zhen Wang
- Department of General Surgery, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang Provincial People's Hospital), Hangzhou Medical College, Taizhou, 317200, Zhejiang, China; Laboratory Medicine Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China.
| | - Ning Hu
- Department of Chemistry, Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 310058, China; General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, China.
| |
Collapse
|
18
|
Turner KM, Patel SH. Pancreatic Cancer Screening among High-risk Individuals. Surg Clin North Am 2024; 104:951-964. [PMID: 39237170 DOI: 10.1016/j.suc.2024.03.002] [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 ductal adenocarcinoma (PDAC) continues to remain one of the leading causes of cancer-related death. Unlike other malignancies where universal screening is recommended, the same cannot be said for PDAC. The purpose of this study is to review which patients are at high risk of developing PDAC and therefore candidates for screening, methods/frequency of screening, and risk for these groups of patients.
Collapse
Affiliation(s)
- Kevin M Turner
- Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0558, USA
| | - Sameer H Patel
- Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0558, USA; Division of Surgical Oncology, Medical Science Building 231 Albert Sabin Way, Cincinnati, OH 45267-0558, USA.
| |
Collapse
|
19
|
Kawai M, Fukuda A, Otomo R, Obata S, Minaga K, Asada M, Umemura A, Uenoyama Y, Hieda N, Morita T, Minami R, Marui S, Yamauchi Y, Nakai Y, Takada Y, Ikuta K, Yoshioka T, Mizukoshi K, Iwane K, Yamakawa G, Namikawa M, Sono M, Nagao M, Maruno T, Nakanishi Y, Hirai M, Kanda N, Shio S, Itani T, Fujii S, Kimura T, Matsumura K, Ohana M, Yazumi S, Kawanami C, Yamashita Y, Marusawa H, Watanabe T, Ito Y, Kudo M, Seno H. Early detection of pancreatic cancer by comprehensive serum miRNA sequencing with automated machine learning. Br J Cancer 2024; 131:1158-1168. [PMID: 39198617 PMCID: PMC11442445 DOI: 10.1038/s41416-024-02794-5] [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: 10/26/2023] [Revised: 06/26/2024] [Accepted: 07/03/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Pancreatic cancer is often diagnosed at advanced stages, and early-stage diagnosis of pancreatic cancer is difficult because of nonspecific symptoms and lack of available biomarkers. METHODS We performed comprehensive serum miRNA sequencing of 212 pancreatic cancer patient samples from 14 hospitals and 213 non-cancerous healthy control samples. We randomly classified the pancreatic cancer and control samples into two cohorts: a training cohort (N = 185) and a validation cohort (N = 240). We created ensemble models that combined automated machine learning with 100 highly expressed miRNAs and their combination with CA19-9 and validated the performance of the models in the independent validation cohort. RESULTS The diagnostic model with the combination of the 100 highly expressed miRNAs and CA19-9 could discriminate pancreatic cancer from non-cancer healthy control with high accuracy (area under the curve (AUC), 0.99; sensitivity, 90%; specificity, 98%). We validated high diagnostic accuracy in an independent asymptomatic early-stage (stage 0-I) pancreatic cancer cohort (AUC:0.97; sensitivity, 67%; specificity, 98%). CONCLUSIONS We demonstrate that the 100 highly expressed miRNAs and their combination with CA19-9 could be biomarkers for the specific and early detection of pancreatic cancer.
Collapse
Affiliation(s)
- Munenori Kawai
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan.
| | - Ryo Otomo
- Research and Development Division, ARKRAY, Inc., Yousuien-nai, 59 Gansuin-cho, Kamigyo-ku, Kyoto, Japan
| | - Shunsuke Obata
- Research and Development Division, ARKRAY, Inc., Yousuien-nai, 59 Gansuin-cho, Kamigyo-ku, Kyoto, Japan
| | - Kosuke Minaga
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Masanori Asada
- Department of Gastroenterology and Hepatology, Osaka Red Cross Hospital, Osaka, Japan
| | - Atsushi Umemura
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Uenoyama
- Department of Gastroenterology and Hepatology, Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
| | - Nobuhiro Hieda
- Department of Gastroenterology, Otsu Red Cross Hospital, Shiga, Japan
| | - Toshihiro Morita
- Department of Gastroenterology and Hepatology, Kitano Hospital, Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Ryuki Minami
- Department of Gastroenterology, Tenri Hospital, Nara, Japan
| | - Saiko Marui
- Department of Gastroenterology and Hepatology, Shiga General Hospital, Shiga, Japan
| | - Yuki Yamauchi
- Department of Gastroenterology, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Japan
| | - Yoshitaka Nakai
- Department of Gastroenterology and Hepatology, Kyoto Katsura Hospital, Kyoto, Japan
| | - Yutaka Takada
- Department of Gastroenterology and Hepatology, Kobe City Nishi-Kobe Medical Center, Kobe, Japan
| | - Kozo Ikuta
- Division of Gastroenterology, Shinko Hospital, Kobe, Japan
| | - Takuto Yoshioka
- Department of Gastroenterology and Hepatology, Takatsuki Red Cross Hospital, Takatsuki, Japan
| | - Kenta Mizukoshi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Kosuke Iwane
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Go Yamakawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Mio Namikawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Makoto Sono
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Munemasa Nagao
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Takahisa Maruno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Yuki Nakanishi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Mitsuharu Hirai
- Research and Development Division, ARKRAY, Inc., Yousuien-nai, 59 Gansuin-cho, Kamigyo-ku, Kyoto, Japan
| | - Naoki Kanda
- Department of Gastroenterology and Hepatology, Takatsuki Red Cross Hospital, Takatsuki, Japan
| | - Seiji Shio
- Division of Gastroenterology, Shinko Hospital, Kobe, Japan
| | - Toshinao Itani
- Department of Gastroenterology and Hepatology, Kobe City Nishi-Kobe Medical Center, Kobe, Japan
| | - Shigehiko Fujii
- Department of Gastroenterology and Hepatology, Kyoto Katsura Hospital, Kyoto, Japan
| | - Toshiyuki Kimura
- Department of Gastroenterology, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Japan
| | - Kazuyoshi Matsumura
- Department of Gastroenterology and Hepatology, Shiga General Hospital, Shiga, Japan
| | - Masaya Ohana
- Department of Gastroenterology, Tenri Hospital, Nara, Japan
| | - Shujiro Yazumi
- Department of Gastroenterology and Hepatology, Kitano Hospital, Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Chiharu Kawanami
- Department of Gastroenterology, Otsu Red Cross Hospital, Shiga, Japan
| | - Yukitaka Yamashita
- Department of Gastroenterology and Hepatology, Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
| | - Hiroyuki Marusawa
- Department of Gastroenterology and Hepatology, Osaka Red Cross Hospital, Osaka, Japan
| | - Tomohiro Watanabe
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yoshito Ito
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
20
|
Seufferlein T, Mayerle J, Boeck S, Brunner T, Ettrich TJ, Grenacher L, Gress TM, Hackert T, Heinemann V, Kestler A, Sinn M, Tannapfel A, Wedding U, Uhl W. S3-Leitlinie Exokrines Pankreaskarzinom – Version 3.1. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:874-995. [PMID: 39389103 DOI: 10.1055/a-2338-3533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Affiliation(s)
| | | | | | - Thomas Brunner
- Universitätsklinik für Strahlentherapie-Radioonkologie, Medizinische Universität Graz, Austria
| | | | | | - Thomas Mathias Gress
- Gastroenterologie und Endokrinologie Universitätsklinikum Gießen und Marburg, Germany
| | - Thilo Hackert
- Klinik und Poliklinik für Allgemein-, Viszeral- und Thoraxchirurgie, Universitätsklinikum Hamburg-Eppendorf, Germany
| | - Volker Heinemann
- Medizinische Klinik und Poliklinik III, Klinikum der Universität München-Campus Grosshadern, München, Germany
| | | | - Marianne Sinn
- Medizinische Klinik und Poliklinik II Onkologie und Hämatologie, Universitätsklinikum Hamburg-Eppendorf, Germany
| | | | | | - Waldemar Uhl
- Allgemein- und Viszeralchirurgie, St Josef-Hospital, Bochum, Germany
| |
Collapse
|
21
|
Cheng HH, Shevach JW, Castro E, Couch FJ, Domchek SM, Eeles RA, Giri VN, Hall MJ, King MC, Lin DW, Loeb S, Morgan TM, Offit K, Pritchard CC, Schaeffer EM, Szymaniak BM, Vassy JL, Katona BW, Maxwell KN. BRCA1, BRCA2, and Associated Cancer Risks and Management for Male Patients: A Review. JAMA Oncol 2024; 10:1272-1281. [PMID: 39052257 PMCID: PMC12080741 DOI: 10.1001/jamaoncol.2024.2185] [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] [Indexed: 07/27/2024]
Abstract
Importance Half of all carriers of inherited cancer-predisposing variants in BRCA1 and BRCA2 are male, but the implications for their health are underrecognized compared to female individuals. Germline variants in BRCA1 and BRCA2 (also known as pathogenic or likely pathogenic variants, referred to here as BRCA1/2 PVs) are well known to significantly increase the risk of breast and ovarian cancers in female carriers, and knowledge of BRCA1/2 PVs informs established cancer screening and options for risk reduction. While risks to male carriers of BRCA1/2 PVs are less characterized, there is convincing evidence of increased risk for prostate cancer, pancreatic cancer, and breast cancer in males. There has also been a rapid expansion of US Food and Drug Administration-approved targeted cancer therapies, including poly ADP ribose polymerase (PARP) inhibitors, for breast, pancreatic, and prostate cancers associated with BRCA1/2 PVs. Observations This narrative review summarized the data that inform cancer risks, targeted cancer therapy options, and guidelines for early cancer detection. It also highlighted areas of emerging research and clinical trial opportunities for male BRCA1/2 PV carriers. These developments, along with the continued relevance to family cancer risk and reproductive options, have informed changes to guideline recommendations for genetic testing and strengthened the case for increased genetic testing for males. Conclusions and Relevance Despite increasing clinical actionability for male carriers of BRCA1/2 PVs, far fewer males than female individuals undergo cancer genetic testing. Oncologists, internists, and primary care clinicians should be vigilant about offering appropriate genetic testing to males. Identifying more male carriers of BRCA1/2 PVs will maximize opportunities for cancer early detection, targeted risk management, and cancer treatment for males, along with facilitating opportunities for risk reduction and prevention in their family members, thereby decreasing the burden of hereditary cancer.
Collapse
Affiliation(s)
- Heather H. Cheng
- Fred Hutchinson Cancer Center, Clinical Research Division, Seattle, WA
- University of Washington, Department of Medicine (Hematology and Oncology), Seattle, WA
| | - Jeffrey W. Shevach
- Duke University School of Medicine, Division of Medical Oncology, Durham, NC
| | - Elena Castro
- Hospital Universitario 12 de Octubre, Department of Medical Oncology, Madrid, Spain
| | - Fergus J. Couch
- Mayo Clinic, Division of Experimental Pathology and Laboratory Medicine, Rochester, NY
| | - Susan M. Domchek
- Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Department of Medicine, Philadelphia, PA
| | - Rosalind A. Eeles
- The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London UK
| | - Veda N. Giri
- Yale School of Medicine and Yale Cancer Center, New Haven, CT
| | - Michael J. Hall
- Fox Chase Cancer Center, Department of Clinical Genetics, Philadelphia, PA
| | - Mary-Claire King
- University of Washington, Department of Medicine (Medical Genetics) and Department of Genome Sciences, Seattle, WA
| | - Daniel W. Lin
- Fred Hutchinson Cancer Center, Clinical Research Division, Seattle, WA
- University of Washington, Department of Urology, Seattle, WA
| | - Stacy Loeb
- New York University School of Medicine, Department of Urology and Population Health, New York, NY
- Manhattan Veterans Affairs, Department of Surgery/Urology, New York, NY
| | - Todd M. Morgan
- University of Michigan, Department of Urology, Ann Arbor, MI
| | - Kenneth Offit
- Memorial Sloan Kettering Cancer Center, Clinical Genetics Service, New York, NY
| | - Colin C. Pritchard
- University of Washington, Department of Laboratory Medicine and Pathology, Seattle, WA
- Brotman Baty Institute for Precision Medicine, Seattle, WA
| | - Edward M. Schaeffer
- Northwestern University, Feinberg School of Medicine, Department of Urology, Chicago, IL
| | - Brittany M. Szymaniak
- Northwestern University, Feinberg School of Medicine, Department of Urology, Chicago, IL
| | - Jason L. Vassy
- Harvard Medical School at VA Boston Healthcare System, Boston, MA
| | - Bryson W. Katona
- Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Department of Medicine, Philadelphia, PA
| | - Kara N. Maxwell
- Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Department of Medicine, Philadelphia, PA
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA
| |
Collapse
|
22
|
Kane WJ, Haden KR, Martin EN, Shami VM, Wang AY, Strand DS, Adair SJ, Nagdas S, Tsung A, Zaydfudim VM, Adams RB, Bauer TW. Survival benefit associated with screening of patients at elevated risk for pancreatic cancer. J Surg Oncol 2024; 130:485-492. [PMID: 39016067 DOI: 10.1002/jso.27784] [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: 06/21/2024] [Accepted: 06/30/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND & OBJECTIVES: Screening for pancreatic cancer is recommended for individuals with a strong family history, certain genetic syndromes, or a neoplastic cyst of the pancreas. However, limited data supports a survival benefit attributable to screening these higher-risk individuals. METHODS All patients enrolled in screening at a High-Risk Pancreatic Cancer Clinic (HRC) from July 2013 to June 2020 were identified from a prospectively maintained institutional database and compared to patients evaluated at a Surgical Oncology Clinic (SOC) at the same institution during the same period. Clinical outcomes of patients selected for surgical resection, particularly clinicopathologic stage and overall survival, were compared. RESULTS Among 826 HRC patients followed for a median (IQR) of 2.3 (0.8-4.2) years, 128 were selected for surgical resection and compared to 402 SOC patients selected for resection. Overall survival was significantly longer among HRC patients (median survival: not reached vs. 2.6 years, p < 0.001). Among 31 HRC and 217 SOC patients with a diagnosis of pancreatic ductal adenocarcinoma (PDAC), the majority of HRC patients were diagnosed with stage 0 disease (carcinoma in situ), while the majority of SOC patients were diagnosed with stage II disease (p < 0.001). Overall survival after resection of invasive PDAC was also significantly longer among HRC patients compared to SOC patients (median survival 5.5 vs. 1.6 years, p = 0.002). CONCLUSION Patients at increased risk for PDAC and followed with guideline-based screening exhibited downstaging of disease and improved survival from PDAC in comparison to patients who were not screened.
Collapse
Affiliation(s)
- William J Kane
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Kathleen R Haden
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Elizabeth N Martin
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Vanessa M Shami
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia, USA
| | - Andrew Y Wang
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel S Strand
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia, USA
| | - Sara J Adair
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Sarbajeet Nagdas
- School of Medicine, University of Virginia, Charlottesville, University of Virginia, USA
| | - Allan Tsung
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Victor M Zaydfudim
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Reid B Adams
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Todd W Bauer
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
23
|
Löhr JM, Öhlund D, Söreskog E, Andersson E, Vujasinovic M, Zethraeus N, Sund M. Can our experience with surveillance for inherited pancreatic cancer help to identify early pancreatic cancer in the general population? Fam Cancer 2024; 23:399-403. [PMID: 38441833 PMCID: PMC11255073 DOI: 10.1007/s10689-024-00363-6] [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] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/05/2024] [Indexed: 07/18/2024]
Abstract
Screening of the general population for cancer is a matter of primary prevention reducing the burden of disease. Whilst this is successful for several cancers including breast, colon and prostate, the situation to screen and hence prevent pancreatic cancer is different. The organ is not as accessible to simple physical exam or biological samples (fecal or blood test). Neither exists a blood test such as PSA that is cost-effective. Reviewing the evidence from screening risk groups for pancreatic cancer, one must conclude that there is no rational at present to screen the general population, for a lack of appropriate tests.
Collapse
Affiliation(s)
- J-Matthias Löhr
- Karolinska Comprehensive Cancer Center and Karolinska Institutet, Stockholm, Sweden.
- Div. of Surgery & Oncology, Dept. of Upper Abdominal Diseases, CLINTEC Karolinska Institutet, Karolinska Comprehensive Cancer Center, Stockholm, SE-141 86, Sweden.
| | - Daniel Öhlund
- Department of Radiation Sciences and Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Emma Söreskog
- Department of Learning, Informatics, Management and Ethics (LIME), Karolinska Institutet, Stockholm, Sweden
| | - Emil Andersson
- Karolinska Comprehensive Cancer Center and Karolinska Institutet, Stockholm, Sweden
| | - Miroslav Vujasinovic
- Karolinska Comprehensive Cancer Center and Karolinska Institutet, Stockholm, Sweden
| | - Niklas Zethraeus
- Department of Learning, Informatics, Management and Ethics (LIME), Karolinska Institutet, Stockholm, Sweden
| | - Malin Sund
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Surgical and Perioperative Sciences/ Surgery, Umeå University, Umeå, Sweden
| |
Collapse
|
24
|
Cristina-Marianini-Rios, Sanchez MEC, de Paredes AGG, Rodríguez M, Barreto E, López JV, Fuentes R, Beltrán MM, Sanjuanbenito A, Lobo E, Caminoa A, Ruz-Caracuel I, Durán SL, Olcina JRF, Blázquez J, Sequeros EV, Carrato A, Ávila JCM, Earl J. The best linear unbiased prediction (BLUP) method as a tool to estimate the lifetime risk of pancreatic ductal adenocarcinoma in high-risk individuals with no known pathogenic germline variants. Fam Cancer 2024; 23:233-246. [PMID: 38780705 PMCID: PMC11254992 DOI: 10.1007/s10689-024-00397-w] [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: 02/02/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related death in the Western world. The number of diagnosed cases and the mortality rate are almost equal as the majority of patients present with advanced disease at diagnosis. Between 4 and 10% of pancreatic cancer cases have an apparent hereditary background, known as hereditary pancreatic cancer (HPC) and familial pancreatic cancer (FPC), when the genetic basis is unknown. Surveillance of high-risk individuals (HRI) from these families by imaging aims to detect PDAC at an early stage to improve prognosis. However, the genetic basis is unknown in the majority of HRIs, with only around 10-13% of families carrying known pathogenic germline mutations. The aim of this study was to assess an individual's genetic cancer risk based on sex and personal and family history of cancer. The Best Linear Unbiased Prediction (BLUP) methodology was used to estimate an individual's predicted risk of developing cancer during their lifetime. The model uses different demographic factors in order to estimate heritability. A reliable estimation of heritability for pancreatic cancer of 0.27 on the liability scale, and 0.07 at the observed data scale as obtained, which is different from zero, indicating a polygenic inheritance pattern of PDAC. BLUP was able to correctly discriminate PDAC cases from healthy individuals and those with other cancer types. Thus, providing an additional tool to assess PDAC risk HRI with an assumed genetic predisposition in the absence of known pathogenic germline mutations.
Collapse
Affiliation(s)
- Cristina-Marianini-Rios
- Department of Agricultural Economics, Statistics and Business Management, Universidad Politécnica de Madrid, Madrid, Spain
| | - María E Castillo Sanchez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
| | - Ana García García de Paredes
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercedes Rodríguez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- University of Alcalá, Madrid, Spain
| | - Emma Barreto
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- University of Alcalá, Madrid, Spain
| | - Jorge Villalón López
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
| | - Raquel Fuentes
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, 28034, Spain
| | | | - Alfonso Sanjuanbenito
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- Pancreatic and Biliopancreatic Surgery Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Eduardo Lobo
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Pancreatic and Biliopancreatic Surgery Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alejandra Caminoa
- Department of Pathology, Hospital Universitario Ramón y Cajal, Madrid, 28034, Spain
| | - Ignacio Ruz-Caracuel
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- Department of Pathology, Hospital Universitario Ramón y Cajal, Madrid, 28034, Spain
| | - Sergio López Durán
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - José Ramón Foruny Olcina
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Javier Blázquez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Radiology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Enrique Vázquez Sequeros
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
| | - Alfredo Carrato
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- University of Alcalá, Madrid, Spain
- Pancreatic Cancer Europe, Brussels, Belgium
| | - Jose Carlos Martínez Ávila
- Department of Agricultural Economics, Statistics and Business Management, Universidad Politécnica de Madrid, Madrid, Spain.
| | - Julie Earl
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain.
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain.
| |
Collapse
|
25
|
Blackford AL, Canto MI, Dbouk M, Hruban RH, Katona BW, Chak A, Brand RE, Syngal S, Farrell J, Kastrinos F, Stoffel EM, Rustgi A, Klein AP, Kamel I, Fishman EK, He J, Burkhart R, Shin EJ, Lennon AM, Goggins M. Pancreatic Cancer Surveillance and Survival of High-Risk Individuals. JAMA Oncol 2024; 10:1087-1096. [PMID: 38959011 PMCID: PMC11223057 DOI: 10.1001/jamaoncol.2024.1930] [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: 10/18/2023] [Accepted: 02/05/2024] [Indexed: 07/04/2024]
Abstract
Importance Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease with increasing incidence. The majority of PDACs are incurable at presentation, but population-based screening is not recommended. Surveillance of high-risk individuals for PDAC may lead to early detection, but the survival benefit is unproven. Objective To compare the survival of patients with surveillance-detected PDAC with US national data. Design, Setting, and Participants This comparative cohort study was conducted in multiple US academic medical centers participating in the Cancer of the Pancreas Screening program, which screens high-risk individuals with a familial or genetic predisposition for PDAC. The comparison cohort comprised patients with PDAC matched for age, sex, and year of diagnosis from the Surveillance, Epidemiology, and End Results (SEER) program. The Cancer of the Pancreas Screening program originated in 1998, and data collection was done through 2021. The data analysis was performed from April 29, 2022, through April 10, 2023. Exposures Endoscopic ultrasonography or magnetic resonance imaging performed annually and standard-of-care surgical and/or oncologic treatment. Main Outcomes and Measures Stage of PDAC at diagnosis, overall survival (OS), and PDAC mortality were compared using descriptive statistics and conditional logistic regression, Cox proportional hazards regression, and competing risk regression models. Sensitivity analyses and adjustment for lead-time bias were also conducted. Results A total of 26 high-risk individuals (mean [SD] age at diagnosis, 65.8 [9.5] years; 15 female [57.7%]) with PDAC were compared with 1504 SEER control patients with PDAC (mean [SD] age at diagnosis, 66.8 [7.9] years; 771 female [51.3%]). The median primary tumor diameter of the 26 high-risk individuals was smaller than in the control patients (2.5 [range, 0.6-5.0] vs 3.6 [range, 0.2-8.0] cm, respectively; P < .001). The high-risk individuals were more likely to be diagnosed with a lower stage (stage I, 10 [38.5%]; stage II, 8 [30.8%]) than matched control patients (stage I, 155 [10.3%]; stage II, 377 [25.1%]; P < .001). The PDAC mortality rate at 5 years was lower for high-risk individuals than control patients (43% vs 86%; hazard ratio, 3.58; 95% CI, 2.01-6.39; P < .001), and high-risk individuals lived longer than matched control patients (median OS, 61.7 [range, 1.9-147.3] vs 8.0 [range, 1.0-131.0] months; 5-year OS rate, 50% [95% CI, 32%-80%] vs 9% [95% CI, 7%-11%]). Conclusions and Relevance These findings suggest that surveillance of high-risk individuals may lead to detection of smaller, lower-stage PDACs and improved survival.
Collapse
Affiliation(s)
- Amanda L. Blackford
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Marcia Irene Canto
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Mohamad Dbouk
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Ralph H. Hruban
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Bryson W. Katona
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Amitabh Chak
- Division of Gastroenterology and Liver Disease, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio
| | - Randall E. Brand
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center, Pennsylvania
| | - Sapna Syngal
- Cancer Genetics and Prevention, Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Gastroenterology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - James Farrell
- Yale Center for Pancreatic Disease, Section of Digestive Disease, Yale University, New Haven, Connecticut
| | - Fay Kastrinos
- Division of Digestive and Liver Diseases, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Elena M. Stoffel
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Anil Rustgi
- Division of Digestive and Liver Diseases, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Alison P. Klein
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Ihab Kamel
- Department of Radiology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Elliot K. Fishman
- Department of Radiology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Jin He
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Richard Burkhart
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Eun Ji Shin
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Anne Marie Lennon
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Radiology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Michael Goggins
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| |
Collapse
|
26
|
Maurer E, Bartsch DK. Surgical aspects related to hereditary pancreatic cancer. Fam Cancer 2024; 23:341-350. [PMID: 38662263 PMCID: PMC11254980 DOI: 10.1007/s10689-024-00384-1] [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: 12/06/2023] [Accepted: 03/29/2024] [Indexed: 04/26/2024]
Abstract
The goal of surveillance programs for individuals at risk (IAR) from familial pancreatic cancer (FPC) families or families with other inherited tumor syndromes predisposing to the development of pancreatic adenocarcinoma (PDAC), such as hereditary pancreatitis or Peutz-Jeghers syndrome, is the dectection and consecutive curative resection of early PDAC or even better its high-grade precursor lesions. Although the indication for surgery is quite established, the extent of surgery is not well defined due to the lack of evidence-based data. In addition, multiple factors have to be taken into account to determine an optimal personalized surgical strategy. This holds especially true since pancreatic surgery is associated with a relatively high morbidity and might impair the quality of life significantly. In this article the surgical aspects in the setting of hereditary PDAC are discussed.
Collapse
Affiliation(s)
- Elisabeth Maurer
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University Marburg, 35043, Baldingerstrasse, Marburg, Germany.
| | - Detlef K Bartsch
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University Marburg, 35043, Baldingerstrasse, Marburg, Germany
| |
Collapse
|
27
|
Boekestijn B, Feshtali S, Vasen H, van Leerdam ME, Bonsing BA, Mieog JSD, Wasser MN. Screening for pancreatic cancer in high-risk individuals using MRI: optimization of scan techniques to detect small lesions. Fam Cancer 2024; 23:295-308. [PMID: 38733421 PMCID: PMC11254973 DOI: 10.1007/s10689-024-00394-z] [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: 11/19/2023] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
Pancreatic cancer has a dismal prognosis in the general population. However, early detection and treatment of disease in high-risk individuals can improve survival, as patients with localized disease and especially patients with lesions smaller than 10 mm show greatly improved 5-year survival rates. To achieve early detection through MRI surveillance programs, optimization of imaging is required. Advances in MRI technologies in both hardware and software over the years have enabled reliable detection of pancreatic cancer at a small size and early stage. Standardization of dedicated imaging protocols for the pancreas are still lacking. In this review we discuss state of the art scan techniques, sequences, reduction of artifacts and imaging strategies that enable early detection of lesions. Furthermore, we present the imaging features of small pancreatic cancers from a large cohort of high-risk individuals. Refinement of MRI techniques, increased scan quality and the use of artificial intelligence may further improve early detection and the prognosis of pancreatic cancer in a screening setting.
Collapse
Affiliation(s)
- Bas Boekestijn
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Shirin Feshtali
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans Vasen
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin N Wasser
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
28
|
Overbeek KA, Cahen DL, Bruno MJ. The role of endoscopic ultrasound in the detection of pancreatic lesions in high-risk individuals. Fam Cancer 2024; 23:279-293. [PMID: 38573399 PMCID: PMC11255057 DOI: 10.1007/s10689-024-00380-5] [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: 12/07/2023] [Accepted: 03/17/2024] [Indexed: 04/05/2024]
Abstract
Individuals at high risk of developing pancreatic ductal adenocarcinoma are eligible for surveillance within research programs. These programs employ periodic imaging in the form of magnetic resonance imaging/magnetic resonance cholangiopancreatography or endoscopic ultrasound for the detection of early cancer or high-grade precursor lesions. This narrative review discusses the role of endoscopic ultrasound within these surveillance programs. It details its overall strengths and limitations, yield, burden on patients, and how it compares to magnetic resonance imaging. Finally, recommendations are given when and how to incorporate endoscopic ultrasound in the surveillance of high-risk individuals.
Collapse
Affiliation(s)
- Kasper A Overbeek
- Erasmus MC Cancer Institute, Department of Gastroenterology & Hepatology, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Djuna L Cahen
- Erasmus MC Cancer Institute, Department of Gastroenterology & Hepatology, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marco J Bruno
- Erasmus MC Cancer Institute, Department of Gastroenterology & Hepatology, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
29
|
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: 0] [Impact Index Per Article: 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
|
30
|
Earl J, Fuentes R, Sanchez MEC, de Paredes AGG, Muñoz M, Sanjuanbenito A, Lobo E, Caminoa A, Rodríguez M, Barreto E, López JV, Ruz-Caracuel I, Durán SL, Olcina JRF, Sánchez BL, Páez SC, Torres A, Blázquez J, Sequeros EV, Carrato A. The Spanish Familial Pancreatic Cancer Registry (PANGENFAM): a decade follow-up of individuals at high-risk for pancreatic cancer. Fam Cancer 2024; 23:383-392. [PMID: 38753287 PMCID: PMC11254983 DOI: 10.1007/s10689-024-00388-x] [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: 12/30/2023] [Accepted: 04/04/2024] [Indexed: 07/18/2024]
Abstract
The Spanish Familial Pancreatic Cancer Registry (PANGENFAM) was established in 2009 and aims to characterize the genotype and phenotype of familial pancreatic cancer (FPC). Furthermore, an early detection screening program for pancreatic ductal adenocarcinoma (PDAC) is provided to healthy high-risk individuals from FPC and hereditary pancreatic cancer families (first-degree relatives). This article describes our experience over the last 10 years in high-risk screening. Hereditary and familial pancreatic cancer families were identified through the oncology and gastroenterology units. High-risk individuals underwent annual screening with endoscopic ultrasound (EUS) and magnetic resonance (MRI) from age 40 or 10 years younger than the youngest affected family member. Results: PANGENFAM has enrolled 290 individuals from 143 families, including 52 PDAC cases and 238 high-risk individuals. All high-risk individuals eligible for screening were offered to enter the surveillance program, with 143 currently participating. Pancreatic abnormalities were detected in 94 individuals (median age 53 years (29-83), with common findings including cystic lesions and inhomogeneous parenchyma. Imaging test concordance was 66%. Surgical intervention was performed in 4 high-risk individuals following highly suspicious lesions detected by imaging. PANGENFAM is a valuable resource for science innovation, such as biobanking, with clinical and imaging data available for analysis. For high-risk families, it may offer a potential for early diagnosis. Collaboration with other national and international registries is needed to increase our understanding of the disease biology and to standardize criteria for inclusion and follow-up, optimizing cost-effectiveness and efficacy.
Collapse
Affiliation(s)
- Julie Earl
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain.
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain.
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain.
| | - Raquel Fuentes
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
| | - María E Castillo Sanchez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
| | - Ana García García de Paredes
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - María Muñoz
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Radiology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alfonso Sanjuanbenito
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Pancreatic and Biliopancreatic Surgery Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Eduardo Lobo
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Pancreatic and Biliopancreatic Surgery Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alejandra Caminoa
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Department of Pathology, Hospital Universitario Ramón y Cajal, 28034, Madrid, Spain
| | - Mercedes Rodríguez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
| | - Emma Barreto
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- University of Alcalá, Madrid, Spain
| | - Jorge Villalón López
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Ignacio Ruz-Caracuel
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- Department of Pathology, Hospital Universitario Ramón y Cajal, 28034, Madrid, Spain
| | - Sergio López Durán
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - José Ramón Foruny Olcina
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Bárbara Luna Sánchez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Sonia Camaño Páez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Ana Torres
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Javier Blázquez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Enrique Vázquez Sequeros
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
| | - Alfredo Carrato
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- University of Alcalá, Madrid, Spain
- Pancreatic Cancer Europe, Brussels, Belgium
| |
Collapse
|
31
|
Huang C, Hecht EM, Soloff EV, Tiwari HA, Bhosale PR, Dasayam A, Galgano SJ, Kambadakone A, Kulkarni NM, Le O, Liau J, Luk L, Rosenthal MH, Sangster GP, Goenka AH. Imaging for Early Detection of Pancreatic Ductal Adenocarcinoma: Updates and Challenges in the Implementation of Screening and Surveillance Programs. AJR Am J Roentgenol 2024; 223:e2431151. [PMID: 38809122 DOI: 10.2214/ajr.24.31151] [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: 05/30/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is one of the most aggressive cancers. It has a poor 5-year survival rate of 12%, partly because most cases are diagnosed at advanced stages, precluding curative surgical resection. Early-stage PDA has significantly better prognoses due to increased potential for curative interventions, making early detection of PDA critically important to improved patient outcomes. We examine current and evolving early detection concepts, screening strategies, diagnostic yields among high-risk individuals, controversies, and limitations of standard-of-care imaging.
Collapse
Affiliation(s)
- Chenchan Huang
- Department of Radiology, NYU Langone Health, 660 First Ave, 3rd Fl, New York, NY 10016
| | | | - Erik V Soloff
- Department of Radiology, University of Washington, Seattle, WA
| | - Hina Arif Tiwari
- Department of Radiology, University of Arizona College of Medicine, Banner University Medicine, Tucson, AZ
| | - Priya R Bhosale
- Department of Radiology, The University of Texas MD Anderson Cancer Center, Bellaire, TX
| | - Anil Dasayam
- Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Samuel J Galgano
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | | | - Naveen M Kulkarni
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI
| | - Ott Le
- Department of Radiology, The University of Texas MD Anderson Cancer Center, Bellaire, TX
| | - Joy Liau
- Department of Radiology, University of California at San Diego, San Diego, CA
| | - Lyndon Luk
- Department of Radiology, Columbia University Medical Center, New York, NY
| | - Michael H Rosenthal
- Department of Radiology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | | |
Collapse
|
32
|
Paiella S, Capurso G, Carrara S, Secchettin E, Casciani F, Frigerio I, Zerbi A, Archibugi L, Bonifacio C, Malleo G, Cavestro GM, Barile M, Larghi A, Assisi D, Fantin A, Milanetto AC, Fabbri C, Casadei R, Donato G, Sassatelli R, De Marchi G, Di Matteo FM, Arcangeli V, Panzuto F, Puzzono M, Dal Buono A, Pezzilli R, Salvia R, Rizzatti G, Casadio M, Franco M, Butturini G, Pasquali C, Coluccio C, Ricci C, Cicchese N, Sereni G, de Pretis N, Stigliano S, Rudnas B, Marasco M, Lionetto G, Arcidiacono PG, Terrin M, Crovetto A, Mannucci A, Laghi L, Bassi C, Falconi M. Outcomes of a 3-Year Prospective Surveillance in Individuals at High Risk of Pancreatic Cancer. Am J Gastroenterol 2024; 119:739-747. [PMID: 37787643 DOI: 10.14309/ajg.0000000000002546] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 09/22/2023] [Indexed: 10/04/2023]
Abstract
INTRODUCTION Pancreatic cancer (PC) surveillance of high-risk individuals (HRI) is becoming more common worldwide, aiming at anticipating PC diagnosis at a preclinical stage. In 2015, the Italian Registry of Families at Risk of Pancreatic Cancer was created. We aimed to assess the prevalence and incidence of pancreatic findings, oncological outcomes, and harms 7 years after the Italian Registry of Families at Risk of Pancreatic Cancer inception, focusing on individuals with at least a 3-year follow-up or developing events before. METHODS HRI (subjects with a family history or mutation carriers with/without a family history were enrolled in 18 centers). They underwent annual magnetic resonance with cholangiopancreatography or endoscopic ultrasound (NCT04095195). RESULTS During the study period (June 2015-September 2022), 679 individuals were enrolled. Of these, 524 (77.2%) underwent at least baseline imaging, and 156 (29.8%) with at least a 3-year follow-up or pancreatic malignancy/premalignancy-related events, and represented the study population. The median age was 51 (interquartile range 16) years. Familial PC cases accounted for 81.4% of HRI and individuals with pathogenic variant for 18.6%. Malignant (n = 8) and premalignant (1 PanIN3) lesions were found in 9 individuals. Five of these 8 cases occurred in pathogenic variant carriers, 4 in familial PC cases (2 tested negative at germline testing and 2 others were not tested). Three of the 8 PC were stage I. Five of the 8 PC were resectable, 3 Stage I, all advanced cases being prevalent. The 1-, 2-, and 3-year cumulative hazard of PC was 1.7%, 2.5%, and 3%, respectively. Median overall and disease-free survival of patients with resected PC were 18 and 12 months (95% CI not computable). Considering HRI who underwent baseline imaging, 6 pancreatic neuroendocrine neoplasms (1 resected) and 1 low-yield surgery (low-grade mixed-intraductal papillary mucinous neoplasm) were also reported. DISCUSSION PC surveillance in a fully public health care system is feasible and safe, and leads to early PC or premalignant lesions diagnoses, mostly at baseline but also over time.
Collapse
Affiliation(s)
- Salvatore Paiella
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Gabriele Capurso
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Centre, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
| | - Silvia Carrara
- Department of Gastroenterology, Endoscopy Unit, Humanitas Research Hospital, IRCCS, Rozzano, Milan, Italy
| | - Erica Secchettin
- Department of Surgery, Dentistry, Pediatrics, and Gynecology, University of Verona, Verona, Italy
| | - Fabio Casciani
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Isabella Frigerio
- Hepatopancreatobiliary Surgery Unit, Pederzoli Hospital, Peschiera del Garda, Verona, Italy
| | - Alessandro Zerbi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Pancreatic Surgery Department, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Livia Archibugi
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Centre, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
| | | | - Giuseppe Malleo
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Giulia Martina Cavestro
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Monica Barile
- Genetics and Cancer Prevention, Humanitas Research Hospital-IRCCS, Milan, Italy
| | - Alberto Larghi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Daniela Assisi
- UOSD Gastroenterologia ed Endoscopia Digestiva Istituto Nazionale Tumori Regina Elena, Rome, Italy
| | - Alberto Fantin
- Gastroenterology Unit Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Anna Caterina Milanetto
- Department of Surgery, Oncology and Gastroenterology, Pancreatic and Endocrine Surgery Unit, University of Padova, Padova, Italy
| | - Carlo Fabbri
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, AUSL Romagna, Italy
| | - Riccardo Casadei
- Division of Pancreatic Surgery, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Giulio Donato
- Gastroenterology Unit, Department of Oncological and Specialty Medicine, Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
| | - Romano Sassatelli
- Unit of Gastroenterology and Digestive Endoscopy, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Giulia De Marchi
- Gastroenterology Unit, Department of Medicine, University of Verona, Verona Italy
| | | | - Valentina Arcangeli
- Romagna Cancer Registry IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori" (IRST), Meldola, Forlì-Cesena, Italy
| | - Francesco Panzuto
- Digestive Disease Unit, Sant' Andrea University Hospital, Rome, Italy
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Marta Puzzono
- Radiology Department, Humanitas Research Hospital-IRCCS, Milan, Italy
| | - Arianna Dal Buono
- Department of Gastroenterology, Endoscopy Unit, Humanitas Research Hospital, IRCCS, Rozzano, Milan, Italy
| | | | - Roberto Salvia
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | | | - Marco Casadio
- UOSD Gastroenterologia ed Endoscopia Digestiva Istituto Nazionale Tumori Regina Elena, Rome, Italy
| | - Monica Franco
- Gastroenterology Unit Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Giovanni Butturini
- Hepatopancreatobiliary Surgery Unit, Pederzoli Hospital, Peschiera del Garda, Verona, Italy
| | - Claudio Pasquali
- Department of Surgery, Oncology and Gastroenterology, Pancreatic and Endocrine Surgery Unit, University of Padova, Padova, Italy
| | - Chiara Coluccio
- Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, AUSL Romagna, Italy
| | - Claudio Ricci
- Division of Pancreatic Surgery, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Internal Medicine and Surgery (DIMEC), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Noemi Cicchese
- Gastroenterology Unit, Department of Oncological and Specialty Medicine, Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
| | - Giuliana Sereni
- Unit of Gastroenterology and Digestive Endoscopy, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Nicolò de Pretis
- Gastroenterology Unit, Department of Medicine, University of Verona, Verona Italy
| | - Serena Stigliano
- Digestive Endoscopy Unit, University Campus Bio-Medico, Rome, Italy
| | - Britt Rudnas
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori "Dino Amadori" (IRST), Meldola, Forlì-Cesena, Italy
| | - Matteo Marasco
- Digestive Disease Unit, Sant' Andrea University Hospital, Rome, Italy
| | - Gabriella Lionetto
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Paolo Giorgio Arcidiacono
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Centre, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
| | - Maria Terrin
- Department of Gastroenterology, Endoscopy Unit, Humanitas Research Hospital, IRCCS, Rozzano, Milan, Italy
| | - Anna Crovetto
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Alessandro Mannucci
- Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luigi Laghi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Claudio Bassi
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Massimo Falconi
- Division of Pancreatic and Transplantation Surgery, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute University, Milan, Italy
| |
Collapse
|
33
|
Wang L, Larki NR, Dobkin J, Salgado S, Ahmad N, Kaplan DE, Yang W, Yang YX. A Clinical Prediction Model to Assess Risk for Pancreatic Cancer Among Patients With Acute Pancreatitis. Pancreas 2024; 53:e254-e259. [PMID: 38266222 PMCID: PMC11214820 DOI: 10.1097/mpa.0000000000002295] [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] [Indexed: 01/26/2024]
Abstract
OBJECTIVES We aimed to develop and validate a prediction model as the first step in a sequential screening strategy to identify acute pancreatitis (AP) individuals at risk for pancreatic cancer (PC). MATERIALS AND METHODS We performed a population-based retrospective cohort study among individuals 40 years or older with a hospitalization for AP in the US Veterans Health Administration. For variable selection, we used least absolute shrinkage and selection operator regression with 10-fold cross-validation to identify a parsimonious logistic regression model for predicting the outcome, PC diagnosed within 2 years after AP. We evaluated model discrimination and calibration. RESULTS Among 51,613 eligible study patients with AP, 801 individuals were diagnosed with PC within 2 years. The final model (area under the receiver operating curve, 0.70; 95% confidence interval, 0.67-0.73) included histories of gallstones, pancreatic cyst, alcohol use, smoking, and levels of bilirubin, triglycerides, alkaline phosphatase, aspartate aminotransferase, alanine aminotransferase, and albumin. If the predicted risk threshold was set at 2% over 2 years, 20.3% of the AP population would undergo definitive screening, identifying nearly 50% of PC associated with AP. CONCLUSIONS We developed a prediction model using widely available clinical factors to identify high-risk patients with PC-associated AP, the first step in a sequential screening strategy.
Collapse
Affiliation(s)
- Louise Wang
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Navid Rahimi Larki
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Jane Dobkin
- Columbia Irving Medical Center, New York City, NY
| | - Sanjay Salgado
- Division of Gastroenterology and Hepatology, New York Presbyterian Hospital/Weill Cornell Medical College, New York, New York, USA
| | - Nuzhat Ahmad
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, Philadelphia, PA
| | - David E. Kaplan
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, Philadelphia, PA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Wei Yang
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, Philadelphia, PA
| | - Yu-Xiao Yang
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, Philadelphia, PA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| |
Collapse
|
34
|
Mukherjee S, Korfiatis P, Patnam NG, Trivedi KH, Karbhari A, Suman G, Fletcher JG, Goenka AH. Assessing the robustness of a machine-learning model for early detection of pancreatic adenocarcinoma (PDA): evaluating resilience to variations in image acquisition and radiomics workflow using image perturbation methods. Abdom Radiol (NY) 2024; 49:964-974. [PMID: 38175255 DOI: 10.1007/s00261-023-04127-1] [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: 06/15/2023] [Revised: 11/08/2023] [Accepted: 11/12/2023] [Indexed: 01/05/2024]
Abstract
PURPOSE To evaluate robustness of a radiomics-based support vector machine (SVM) model for detection of visually occult PDA on pre-diagnostic CTs by simulating common variations in image acquisition and radiomics workflow using image perturbation methods. METHODS Eighteen algorithmically generated-perturbations, which simulated variations in image noise levels (σ, 2σ, 3σ, 5σ), image rotation [both CT image and the corresponding pancreas segmentation mask by 45° and 90° in axial plane], voxel resampling (isotropic and anisotropic), gray-level discretization [bin width (BW) 32 and 64)], and pancreas segmentation (sequential erosions by 3, 4, 6, and 8 pixels and dilations by 3, 4, and 6 pixels from the boundary), were introduced to the original (unperturbed) test subset (n = 128; 45 pre-diagnostic CTs, 83 control CTs with normal pancreas). Radiomic features were extracted from pancreas masks of these additional test subsets, and the model's performance was compared vis-a-vis the unperturbed test subset. RESULTS The model correctly classified 43 out of 45 pre-diagnostic CTs and 75 out of 83 control CTs in the unperturbed test subset, achieving 92.2% accuracy and 0.98 AUC. Model's performance was unaffected by a three-fold increase in noise level except for sensitivity declining to 80% at 3σ (p = 0.02). Performance remained comparable vis-a-vis the unperturbed test subset despite variations in image rotation (p = 0.99), voxel resampling (p = 0.25-0.31), change in gray-level BW to 32 (p = 0.31-0.99), and erosions/dilations up to 4 pixels from the pancreas boundary (p = 0.12-0.34). CONCLUSION The model's high performance for detection of visually occult PDA was robust within a broad range of clinically relevant variations in image acquisition and radiomics workflow.
Collapse
Affiliation(s)
- Sovanlal Mukherjee
- Divisions of Abdominal and Nuclear Imaging, Nuclear Radiology Fellowship, Nuclear Radiology Research Operations, Enterprise PET/MR Research and Development, Department of Radiology, Mayo Clinic, 200 First St SW, Charlton 1, Rochester, MN, 55905, USA
| | - Panagiotis Korfiatis
- Divisions of Abdominal and Nuclear Imaging, Nuclear Radiology Fellowship, Nuclear Radiology Research Operations, Enterprise PET/MR Research and Development, Department of Radiology, Mayo Clinic, 200 First St SW, Charlton 1, Rochester, MN, 55905, USA
| | - Nandakumar G Patnam
- Divisions of Abdominal and Nuclear Imaging, Nuclear Radiology Fellowship, Nuclear Radiology Research Operations, Enterprise PET/MR Research and Development, Department of Radiology, Mayo Clinic, 200 First St SW, Charlton 1, Rochester, MN, 55905, USA
| | - Kamaxi H Trivedi
- Divisions of Abdominal and Nuclear Imaging, Nuclear Radiology Fellowship, Nuclear Radiology Research Operations, Enterprise PET/MR Research and Development, Department of Radiology, Mayo Clinic, 200 First St SW, Charlton 1, Rochester, MN, 55905, USA
| | - Aashna Karbhari
- Divisions of Abdominal and Nuclear Imaging, Nuclear Radiology Fellowship, Nuclear Radiology Research Operations, Enterprise PET/MR Research and Development, Department of Radiology, Mayo Clinic, 200 First St SW, Charlton 1, Rochester, MN, 55905, USA
| | - Garima Suman
- Divisions of Abdominal and Nuclear Imaging, Nuclear Radiology Fellowship, Nuclear Radiology Research Operations, Enterprise PET/MR Research and Development, Department of Radiology, Mayo Clinic, 200 First St SW, Charlton 1, Rochester, MN, 55905, USA
| | - Joel G Fletcher
- Divisions of Abdominal and Nuclear Imaging, Nuclear Radiology Fellowship, Nuclear Radiology Research Operations, Enterprise PET/MR Research and Development, Department of Radiology, Mayo Clinic, 200 First St SW, Charlton 1, Rochester, MN, 55905, USA
| | - Ajit H Goenka
- Divisions of Abdominal and Nuclear Imaging, Nuclear Radiology Fellowship, Nuclear Radiology Research Operations, Enterprise PET/MR Research and Development, Department of Radiology, Mayo Clinic, 200 First St SW, Charlton 1, Rochester, MN, 55905, USA.
| |
Collapse
|
35
|
Scherübl H. [Early detection of sporadic pancreatic cancer]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:412-419. [PMID: 37827502 DOI: 10.1055/a-2114-9847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
The incidence of pancreatic cancer is rising. At present, pancreatic cancer is the third most common cancer-causing death in Germany, but it is expected to become the second in 2030 and finally the leading cause of cancer death in 2050. Pancreatic ductal adenocarcinoma (PC) is generally diagnosed at advanced stages, and 5-year-survival has remained poor. Early detection of sporadic PC at stage IA, however, can yield a 5-year-survival rate of about 80%. Early detection initiatives aim at identifying persons at high risk. People with new-onset diabetes at age 50 or older have attracted much interest. Novel strategies regarding how to detect sporadic PC at an early stage are being discussed.
Collapse
Affiliation(s)
- Hans Scherübl
- Klinik für Innere Medizin; Gastroenterol., GI Onkol. u. Infektiol., Vivantes Klinikum Am Urban, Berlin, Germany
- Akademisches Lehrkrankenhaus der Charité, Berlin, Germany
| |
Collapse
|
36
|
Ikezawa K, Fukuda J, Nakao M, Nakano Y, Higashi C, Chagi M, Nakaya Y, Ohkawa K. Correlation between main pancreatic duct diameter measurements: Special pancreatic ultrasonography versus magnetic resonance cholangiopancreatography. Medicine (Baltimore) 2024; 103:e37283. [PMID: 38394509 PMCID: PMC11309629 DOI: 10.1097/md.0000000000037283] [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: 09/23/2023] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Main pancreatic duct (MPD) dilatation is reported to be a risk factor for pancreatic cancer (PC). Although magnetic resonance cholangiopancreatography (MRCP) and ultrasonographic modalities are valuable for monitoring the pancreas, there is limited information on the efficacy of different imaging modalities in measuring MPD diameter. To improve pancreatic imaging, we developed a specialized ultrasound approach focusing on the pancreas (special pancreatic US). We aimed to examine the correlation between MPD diameter measurements using special pancreatic US versus MRCP. We retrospectively reviewed the clinical data of patients with MPD dilation (≥2.5 mm) via special pancreatic US used for screening at our institution between January 2020 and October 2022 and included patients who underwent magnetic resonance imaging 2 months before and after pancreatic US. The MPD diameter on MRCP was measured at the pancreatic locus, where the maximum MPD diameter was obtained on special pancreatic US. This study included 96 patients, with a median interval of 8.5 days between the date of special pancreatic US and the date of undergoing MRCP. MPD dilatation and/or pancreatic cysts were diagnosed in 86 patients, PC in 5 patients, and other diseases in 5 patients. The median MPD diameter, measured using special pancreatic US, was 3.4 mm (interquartile range: 2.9-4.9 mm), whereas it was 3.5 mm using MRCP (interquartile range: 2.8-4.5 mm). There were strong positive correlations between MPD diameter measured on special pancreatic US and that measured on MRCP (R = 0.925, P < .001). This study revealed strong positive correlations between the MPD diameter measurements using special pancreatic US and MRCP. MPD diameter measurements from each imaging method can be helpful during follow-up in individuals at a high risk of PC.
Collapse
Affiliation(s)
- Kenji Ikezawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Junko Fukuda
- Department of Clinical Laboratory, Osaka International Cancer Institute, Osaka, Japan
| | - Miho Nakao
- Department of Clinical Laboratory, Osaka International Cancer Institute, Osaka, Japan
| | - Yoko Nakano
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Chiaki Higashi
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Mayumi Chagi
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Yasuhiro Nakaya
- Department of Diagnostic and Interventional Radiology, Osaka International Cancer Institute, Osaka, Japan
| | - Kazuyoshi Ohkawa
- Department of Hepatobiliary and Pancreatic Oncology, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
37
|
Gao L, Ugalde A, Livingston PM, White V, Watts JJ, Jongebloed H, McCaffrey N, Menzies D, Robinson S. Simulating the healthcare workforce impact and capacity for pancreatic cancer care in Victoria: a model-based analysis. BMC Health Serv Res 2024; 24:239. [PMID: 38395852 PMCID: PMC10893744 DOI: 10.1186/s12913-024-10722-9] [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: 06/09/2023] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The incidence of pancreatic cancer is rising. With improvements in knowledge for screening and early detection, earlier detection of pancreatic cancer will continue to be more common. To support workforce planning, our aim is to perform a model-based analysis that simulates the potential impact on the healthcare workforce, assuming an earlier diagnosis of pancreatic cancer. METHODS We developed a simulation model to estimate the demand (i.e. new cases of pancreatic cancer) and supply (i.e. the healthcare workforce including general surgeons, medical oncologists, radiation oncologists, pain medicine physicians, and palliative care physicians) between 2023 and 2027 in Victoria, Australia. The model compares the current scenario to one in which pancreatic cancer is diagnosed at an earlier stage. The incidence of pancreatic cancer in Victoria, five-year survival rates, and Victoria's population size were obtained from Victorian Cancer Registry, Cancer Council NSW, and Australian Bureau of Statistics respectively. The healthcare workforce data were sourced from the Australian Government Department of Health and Aged Care's Health Workforce Data. The model was constructed at the remoteness level. We analysed the new cases and the number of healthcare workforce by profession together to assess the impact on the healthcare workforce. RESULTS In the status quo, over the next five years, there will be 198 to 220 stages I-II, 297 to 330 stage III, and 495 to 550 stage IV pancreatic cancer cases diagnosed annually, respectively. Assuming 20-70% of the shift towards pancreatic cancer's earlier diagnosis (shifting from stage IV to stages I-II pancreatic cancer within one year), the stages I-II cases could increase to 351 to 390 or 598 to 665 per year. The shift to early diagnosis led to substantial survival gains, translating into an additional 284 or 795 out of 5246 patients with pancreatic cancer remaining alive up to year 5 post-diagnosis. Workforce supply decreases significantly by the remoteness levels, and remote areas face a shortage of key medical professionals registered in delivering pancreatic cancer care, suggesting travel necessities by patients or clinicians. CONCLUSION Improving the early detection and diagnosis of pancreatic cancer is expected to bring significant survival benefits, although there are workforce distribution imbalances in Victoria that may affect the ability to achieve the anticipated survival gain.
Collapse
Affiliation(s)
- Lan Gao
- Deakin Health Economics, Institute of Health Transformation, Faculty of Health, Deakin University, 1 Gheringhap St, 3220, Geelong, Australia.
| | - Anna Ugalde
- School of Nursing & Midwifery, Institute of Health Transformation, Faculty of Health, Deakin University, Melbourne, Australia
| | - Patricia M Livingston
- School of Nursing & Midwifery, Institute of Health Transformation, Faculty of Health, Deakin University, Melbourne, Australia
| | - Victoria White
- School of Nursing & Midwifery, Institute of Health Transformation, Faculty of Health, Deakin University, Melbourne, Australia
| | - Jennifer J Watts
- Deakin Health Economics, Institute of Health Transformation, Faculty of Health, Deakin University, 1 Gheringhap St, 3220, Geelong, Australia
| | - Hannah Jongebloed
- School of Nursing & Midwifery, Institute of Health Transformation, Faculty of Health, Deakin University, Melbourne, Australia
| | - Nikki McCaffrey
- Deakin Health Economics, Institute of Health Transformation, Faculty of Health, Deakin University, 1 Gheringhap St, 3220, Geelong, Australia
| | | | - Suzanne Robinson
- Deakin Health Economics, Institute of Health Transformation, Faculty of Health, Deakin University, 1 Gheringhap St, 3220, Geelong, Australia
| |
Collapse
|
38
|
Laish I, Schechter M, Dancour A, Lieberman S, Levi Z, Goldberg Y, Kedar I, Hasnis E, Half E, Levi GR, Katz L, Vainer ED, Genzel D, Aharoni M, Chen-Shtoyerman R, Abu-Freha N, Raitses-Gurevich M, Golan T, Bernstein-Molho R, Ben Yehoyada M, Gluck N, Rosner G. The benefit of pancreatic cancer surveillance in carriers of germline BRCA1/2 pathogenic variants. Cancer 2024; 130:256-266. [PMID: 37861363 DOI: 10.1002/cncr.35052] [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: 04/08/2023] [Revised: 07/22/2023] [Accepted: 08/03/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Surveillance of high-risk individuals for pancreatic ductal adenocarcinoma (PDAC) is recommended. This study aimed to determine the prevalence and outcomes of PDAC and its precursor lesions in BRCA1/2 pathogenic variants (PVs) carriers undergoing pancreatic surveillance. METHODS A retrospective multicenter cohort study of pancreatic surveillance outcomes in Israeli BRCA1/2 carriers preferably with a family history of PDAC. RESULTS A total of 180 asymptomatic carriers participated in the screening programs, including 57 (31.7%) with BRCA1 PVs, 121 (67.2%) with BRCA2 PVs, and 12 (6.6%) with PVs in BRCA1/2 and other genes, for a median follow-up period of 4 years. Ninety-one individuals (50.5%) fulfilled the International Cancer of the Pancreas Screening (CAPS) criteria for surveillance whereas 116 (64.4%) fulfilled the American College of Gastroenterology (ACG) criteria. There were four cases of adenocarcinoma and four cases of grade 1-neuroendocrine tumor (G1-NET). All were BRCA2 carriers, and two had no family history of PDAC. Three cancer patients were at resectable stages (IA, IIA, IIB) whereas one had a stage IIIB tumor. Of the G1-NET cases, one had surgery and the others were only followed. Success rate for detection of confined pancreatic carcinoma was thus 1.6% (three of 180) in the whole cohort, 1.6% (two of 116) among individuals who fulfilled ACG criteria and 2.2% (two of 91) in those fulfilling CAPS criteria for surveillance. CONCLUSIONS Despite the low detection rate of PDAC and its' high-risk neoplastic precursor lesions among BRCA1/2 carriers undergoing pancreatic surveillance, 75% of cancer cases were detected at a resectable stage.
Collapse
Affiliation(s)
- Ido Laish
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Menachem Schechter
- Gastroenterology Institute, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Alain Dancour
- Gastroenterology Institute, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Sari Lieberman
- Medical Genetics Institute, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Zohar Levi
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Gastroenterology Institute, Beilinson Hospital, Petah Tikva, Israel
| | - Yael Goldberg
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Raphael Recanati Genetics Institute, Beilinson Hospital, Petah Tikva, Israel
| | - Inbal Kedar
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Raphael Recanati Genetics Institute, Beilinson Hospital, Petah Tikva, Israel
| | - Erez Hasnis
- Gastroenterology Institute, Rambam Health Care Campus, Haifa, Israel
| | - Elizabeth Half
- Gastroenterology Institute, Rambam Health Care Campus, Haifa, Israel
| | | | - Lior Katz
- Section of Gastroenterology and Hepatology, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Elez D Vainer
- Section of Gastroenterology and Hepatology, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Dor Genzel
- Section of Gastroenterology and Hepatology, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Maya Aharoni
- Gastroenterology Institute, Kaplan Medical Center, Rehovot, Israel
| | - Rakefet Chen-Shtoyerman
- The Genetic Institute, Kaplan Medical Center, Rehovot, Israel
- The Adelson School of Medicine and the Molecular Biology Department, Ariel University, Ariel, Israel
| | - Naim Abu-Freha
- The Institute of Gastroenterology and Hepatology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Maria Raitses-Gurevich
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Talia Golan
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Rinat Bernstein-Molho
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Danek Gertner Institute of Human Genetics, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Merav Ben Yehoyada
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Nathan Gluck
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Guy Rosner
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| |
Collapse
|
39
|
Das KK. Intraductal Papillary Mucinous Neoplasms in High-Risk Individuals: True, True, and Related? Clin Gastroenterol Hepatol 2024; 22:20-21. [PMID: 37442319 DOI: 10.1016/j.cgh.2023.06.027] [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] [Received: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023]
Affiliation(s)
- Koushik K Das
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
40
|
Overbeek KA, Koopmann BDM, Levink IJM, Tacelli M, Erler NS, Arcidiacono PG, Ausems MGE, Wagner A, van Eijck CH, Groot Koerkamp B, Busch OR, Besselink MG, van der Vlugt M, van Driel LMJW, Fockens P, Vleggaar FP, Poley JW, Capurso G, Cahen DL, Bruno MJ. Intraductal Papillary Mucinous Neoplasms in High-Risk Individuals: Incidence, Growth Rate, and Malignancy Risk. Clin Gastroenterol Hepatol 2024; 22:62-71.e7. [PMID: 37031711 DOI: 10.1016/j.cgh.2023.03.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/19/2023] [Accepted: 03/29/2023] [Indexed: 04/11/2023]
Abstract
BACKGROUND AND AIMS In high-risk individuals (HRIs), we aimed to assess the cumulative incidence of intraductal papillary mucinous neoplasms (IPMNs) and compare IPMN growth, neoplastic progression rate, and the value of growth as predictor for neoplastic progression to these in sporadic IPMNs. METHODS We performed annual surveillance of Dutch HRIs, involving carriers of germline pathogenic variants (PVs) and PV-negative familial pancreatic cancer kindreds. HRIs with IPMNs were compared with Italian individuals without familial risk under surveillance for sporadic IPMNs. RESULTS A total of 457 HRIs were followed for 48 (range 2-172) months; the estimated cumulative IPMN incidence was 46% (95% confidence interval, 28%-64%). In comparison with 442 control individuals, IPMNs in HRIs were more likely to grow ≥2.5 mm/y (31% vs 7%; P < .001) and develop worrisome features (32% vs 19%; P = .010). PV carriers with IPMNs more often displayed neoplastic progression (n = 3 [11%] vs n = 6 [1%]; P = .011), while familial pancreatic cancer kindreds did not (n = 0 [0%]; P = 1.000). The malignancy risk in a PV carrier with an IPMN was 23% for growth rates ≥2.5 mm/y (n = 13), 30% for ≥5 mm/y (n = 10), and 60% for ≥10 mm/y (n = 5). CONCLUSIONS The cumulative incidence of IPMNs in HRIs is higher than previously reported in the general population. Compared with sporadic IPMNs, they have an increased growth rate. PV carriers with IPMNs are suggested to be at a higher malignancy risk. Intensive follow-up should be considered for PV carriers with an IPMN growing ≥2.5 mm/y, and surgical resection for those growing ≥5 mm/y.
Collapse
Affiliation(s)
- Kasper A Overbeek
- Department of Gastroenterology and Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Brechtje D M Koopmann
- Department of Gastroenterology and Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Iris J M Levink
- Department of Gastroenterology and Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Matteo Tacelli
- Pancreato-Biliary Endoscopy and Endoscopic Ultrasound, Pancreas Translational and Clinical Research Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicole S Erler
- Department of Biostatistics, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Paolo Giorgio Arcidiacono
- Pancreato-Biliary Endoscopy and Endoscopic Ultrasound, Pancreas Translational and Clinical Research Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Margreet G E Ausems
- Division Laboratories, Pharmacy and Biomedical Genetics, Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Anja Wagner
- Department of Clinical Genetics, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Casper H van Eijck
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Olivier R Busch
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Marc G Besselink
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Manon van der Vlugt
- Department of Gastroenterology & Hepatology, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology & Metabolism, University of Amsterdam, Amsterdam, the Netherlands
| | - Lydi M J W van Driel
- Department of Gastroenterology and Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Paul Fockens
- Department of Gastroenterology & Hepatology, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology & Metabolism, University of Amsterdam, Amsterdam, the Netherlands
| | - Frank P Vleggaar
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jan-Werner Poley
- Department of Gastroenterology and Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endoscopic Ultrasound, Pancreas Translational and Clinical Research Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy; Digestive and Liver Disease Unit, Sant'Andrea Hospital, Rome, Italy
| | - Djuna L Cahen
- Department of Gastroenterology and Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marco J Bruno
- Department of Gastroenterology and Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
41
|
Paiella S, Secchettin E, Lionetto G, Archibugi L, Azzolina D, Casciani F, Simeone DM, Overbeek KA, Goggins M, Farrell J, Ponz de Leon Pisani R, Tridenti M, Corciulo MA, Malleo G, Arcidiacono PG, Falconi M, Gregori D, Bassi C, Salvia R, Capurso G. Surveillance of Individuals at High Risk of Developing Pancreatic Cancer: A Prevalence Meta-analysis to Estimate the Rate of Low-yield Surgery. Ann Surg 2024; 279:37-44. [PMID: 37681303 DOI: 10.1097/sla.0000000000006094] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
OBJECTIVE To quantify the rate of low-yield surgery, defined as no high-grade dysplastic precursor lesions or T1N0M0 pancreatic cancer at pathology, during pancreatic cancer surveillance. BACKGROUND Global efforts have been made in pancreatic cancer surveillance to anticipate the diagnosis of pancreatic cancer at an early stage and improve survival in high-risk individuals (HRIs) with a hereditary predisposition. The negative impact of pancreatic cancer surveillance when surgery is performed for low-grade dysplasia or a non-neoplastic condition is not well quantified. MATERIALS AND METHODS A systematic search and prevalence meta-analysis was performed for studies reporting surgery with final diagnoses other than those defined by the Cancer of the Pancreas Screening (CAPS) goals from January 2000 to July 2023. The secondary outcome was the pooled proportion of final diagnoses matching the CAPS goals (PROSPERO: #CRD42022300408). RESULTS Twenty-three articles with 5027 patients (median 109 patients/study, interquartile range 251) were included. The pooled prevalence of low-yield surgery was 2.1% (95% CI: 0.9-3.7, I2 : 83%). In the subgroup analysis, this prevalence was nonsignificantly higher in studies that only included familial pancreatic cancer subjects without known pathogenic variants, compared with those enrolling pathogenic variant carriers. No effect modifiers were found. Overall, the pooled prevalence of subjects under surveillance who had a pancreatic resection that contained target lesions was 0.8% (95% CI, 0.3-1.5, I2 : 24%]. The temporal analysis showed that the rate of low-yield surgeries decreased in the last decades and stabilized at around 1% (test for subgroup differences P <0.01). CONCLUSIONS The risk of "low-yield" surgery during pancreatic cancer surveillance is relatively low but should be thoroughly discussed with individuals under surveillance.
Collapse
Affiliation(s)
- Salvatore Paiella
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Erica Secchettin
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Gabriella Lionetto
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Livia Archibugi
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| | - Danila Azzolina
- Department of Environmental and Preventive Science, University of Ferrara, Ferrara, Italy
| | - Fabio Casciani
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Diane M Simeone
- Department of Surgery, New York University, New York, NY
- Perlmutter Cancer Center, New York University, New York, NY
| | - Kasper A Overbeek
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Michael Goggins
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - James Farrell
- Yale Center for Pancreatic Disease, Yale University School of Medicine, New Haven, CT
| | - Ruggero Ponz de Leon Pisani
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| | - Maddalena Tridenti
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| | - Maria Assunta Corciulo
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, Padova, Italy
| | - Giuseppe Malleo
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Paolo Giorgio Arcidiacono
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| | - Massimo Falconi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Pancreatic Surgery and Transplantation Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Dario Gregori
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, Padova, Italy
| | - Claudio Bassi
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Roberto Salvia
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| |
Collapse
|
42
|
Wu BU, Chen Q, Moon BH, Lustigova E, Nielsen EG, Alvarado M, Ahmed SA. Association of Glycated Hemoglobin With a Risk of Pancreatic Cancer in High-Risk Individuals Based on Genetic and Family History. Clin Transl Gastroenterol 2024; 15:e00650. [PMID: 37800692 PMCID: PMC10810597 DOI: 10.14309/ctg.0000000000000650] [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: 06/05/2023] [Accepted: 09/21/2023] [Indexed: 10/07/2023] Open
Abstract
INTRODUCTION Screening for pancreatic cancer (PC) is suggested for high-risk individuals. Additional risk factors may enhance early detection in this population. METHODS Retrospective cohort study among patients with germline variants and/or familial pancreatic cancer in an integrated healthcare system between 2003 and 2019. We calculated the incidence rate (IR) by risk category and performed a nested case-control study to evaluate the relationship between HbA1C and PC within 3 years before diagnosis (cases) or match date (controls). Cases were matched 1:4 by age, sex, and timing of HbA1c. Logistic regression was performed to assess an independent association with PC. RESULTS We identified 5,931 high-risk individuals: 1,175(19.8%) familial PC, 45(0.8%) high-risk germline variants ( STK11, CDKN2A ), 4,097(69.1%) had other germline variants ( ATM, BRCA 1, BRCA 2, CASR, CDKN2A, CFTR, EPCAM, MLH1, MSH2, MSH6, PALB2, PRSS1, STK11, and TP53 ), and 614(10.4%) had both germline variants and family history. Sixty-eight patients (1.1%) developed PC; 50% were metastatic at diagnosis. High-risk variant was associated with greatest risk of PC, IR = 85.1(95% confidence interval: 36.7-197.6)/10,000 person-years; other germline variants and first-degree relative had IR = 33 (18.4, 59.3), whereas IR among ≥2 first-degree relative alone was 10.7 (6.1, 18.8). HbA1c was significantly higher among cases vs controls (median = 7.0% vs 6.4%, P = 0.02). In multivariable analysis, every 1% increase in HbA1c was associated with 36% increase in odds of PC (odds ratio 1.36, 95% confidence interval: 1.08-1.72). Pancreatitis was independently associated with a risk of PC (odds ratio 3.93, 95% confidence limit 1.19, 12.91). DISCUSSION Risk of PC varies among high-risk individuals. HbA1c and history of pancreatitis may be useful additional markers for early detection in this patient population.
Collapse
Affiliation(s)
- Bechien U. Wu
- Center for Pancreatic Care, Division of Gastroenterology, Kaiser Permanente Los Angeles Medical Center, Los Angeles, California, USA;
| | - Qiaoling Chen
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California, USA;
| | - Becky H. Moon
- Center for Pancreatic Care, Division of Gastroenterology, Kaiser Permanente Los Angeles Medical Center, Los Angeles, California, USA;
| | - Eva Lustigova
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California, USA;
| | - Erin G. Nielsen
- Department of Genetics, Southern California Medical Group, Pasadena, California, USA;
| | - Monica Alvarado
- Department of Genetics, Southern California Medical Group, Pasadena, California, USA;
| | - Syed A. Ahmed
- Department of Genetics, Kaiser Permanente Riverside Medical Center, Riverside, California, USA
| |
Collapse
|
43
|
Abstract
Pancreatic cancer remains among the malignancies with the worst outcomes. Survival has been improving, but at a slower rate than other cancers. Multimodal treatment, including chemotherapy, surgical resection, and radiotherapy, has been under investigation for many years. Because of the anatomical characteristics of the pancreas, more emphasis on treatment selection has been placed on local extension into major vessels. Recently, the development of more effective treatment regimens has opened up new treatment strategies, but urgent research questions have also become apparent. This review outlines the current management of pancreatic cancer, and the recent advances in its treatment. The review discusses future treatment pathways aimed at integrating novel findings of translational and clinical research.
Collapse
Affiliation(s)
- Marco Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, USA
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Toshitaka Sugawara
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sana D Karam
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Wells A Messersmith
- University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, CO, USA
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
44
|
Korfiatis P, Suman G, Patnam NG, Trivedi KH, Karbhari A, Mukherjee S, Cook C, Klug JR, Patra A, Khasawneh H, Rajamohan N, Fletcher JG, Truty MJ, Majumder S, Bolan CW, Sandrasegaran K, Chari ST, Goenka AH. Automated Artificial Intelligence Model Trained on a Large Data Set Can Detect Pancreas Cancer on Diagnostic Computed Tomography Scans As Well As Visually Occult Preinvasive Cancer on Prediagnostic Computed Tomography Scans. Gastroenterology 2023; 165:1533-1546.e4. [PMID: 37657758 PMCID: PMC10843414 DOI: 10.1053/j.gastro.2023.08.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND & AIMS The aims of our case-control study were (1) to develop an automated 3-dimensional (3D) Convolutional Neural Network (CNN) for detection of pancreatic ductal adenocarcinoma (PDA) on diagnostic computed tomography scans (CTs), (2) evaluate its generalizability on multi-institutional public data sets, (3) its utility as a potential screening tool using a simulated cohort with high pretest probability, and (4) its ability to detect visually occult preinvasive cancer on prediagnostic CTs. METHODS A 3D-CNN classification system was trained using algorithmically generated bounding boxes and pancreatic masks on a curated data set of 696 portal phase diagnostic CTs with PDA and 1080 control images with a nonneoplastic pancreas. The model was evaluated on (1) an intramural hold-out test subset (409 CTs with PDA, 829 controls); (2) a simulated cohort with a case-control distribution that matched the risk of PDA in glycemically defined new-onset diabetes, and Enriching New-Onset Diabetes for Pancreatic Cancer score ≥3; (3) multi-institutional public data sets (194 CTs with PDA, 80 controls), and (4) a cohort of 100 prediagnostic CTs (i.e., CTs incidentally acquired 3-36 months before clinical diagnosis of PDA) without a focal mass, and 134 controls. RESULTS Of the CTs in the intramural test subset, 798 (64%) were from other hospitals. The model correctly classified 360 CTs (88%) with PDA and 783 control CTs (94%), with a mean accuracy 0.92 (95% CI, 0.91-0.94), area under the receiver operating characteristic (AUROC) curve of 0.97 (95% CI, 0.96-0.98), sensitivity of 0.88 (95% CI, 0.85-0.91), and specificity of 0.95 (95% CI, 0.93-0.96). Activation areas on heat maps overlapped with the tumor in 350 of 360 CTs (97%). Performance was high across tumor stages (sensitivity of 0.80, 0.87, 0.95, and 1.0 on T1 through T4 stages, respectively), comparable for hypodense vs isodense tumors (sensitivity: 0.90 vs 0.82), different age, sex, CT slice thicknesses, and vendors (all P > .05), and generalizable on both the simulated cohort (accuracy, 0.95 [95% 0.94-0.95]; AUROC curve, 0.97 [95% CI, 0.94-0.99]) and public data sets (accuracy, 0.86 [95% CI, 0.82-0.90]; AUROC curve, 0.90 [95% CI, 0.86-0.95]). Despite being exclusively trained on diagnostic CTs with larger tumors, the model could detect occult PDA on prediagnostic CTs (accuracy, 0.84 [95% CI, 0.79-0.88]; AUROC curve, 0.91 [95% CI, 0.86-0.94]; sensitivity, 0.75 [95% CI, 0.67-0.84]; and specificity, 0.90 [95% CI, 0.85-0.95]) at a median 475 days (range, 93-1082 days) before clinical diagnosis. CONCLUSIONS This automated artificial intelligence model trained on a large and diverse data set shows high accuracy and generalizable performance for detection of PDA on diagnostic CTs as well as for visually occult PDA on prediagnostic CTs. Prospective validation with blood-based biomarkers is warranted to assess the potential for early detection of sporadic PDA in high-risk individuals.
Collapse
Affiliation(s)
| | - Garima Suman
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | - Cole Cook
- Division of Medical Imaging Technology Services, Mayo Clinic, Rochester, Minnesota
| | - Jason R Klug
- Division of Medical Imaging Technology Services, Mayo Clinic, Rochester, Minnesota
| | - Anurima Patra
- Department of Radiology, Tata Medical Center, Kolkata, India
| | - Hala Khasawneh
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Mark J Truty
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Shounak Majumder
- Department of Gastroenterology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Suresh T Chari
- Department of Gastroenterology, Mayo Clinic, Rochester, Minnesota
| | - Ajit H Goenka
- Department of Radiology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
45
|
Sirtl S, Vornhülz M, Hofmann FO, Mayerle J, Beyer G. [Pancreatic cancer-screening or surveillance?]. RADIOLOGIE (HEIDELBERG, GERMANY) 2023; 63:908-915. [PMID: 37878016 DOI: 10.1007/s00117-023-01227-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
BACKGROUND Despite continuous improvement of diagnostic and therapeutic procedures, the number of new pancreatic ductal adenocarcinoma (PDAC) cases diagnosed annually almost equals the number of PDAC-related deaths. Prerequisite for curative treatment is a resectable tumor at the time of diagnosis. Individuals with genetic and/or familial risk profiles should therefore be screened and included in structured surveillance programs. OBJECTIVES Description of the status quo and usefulness of current PDAC screening and surveillance concepts. METHODS A selective literature search of current national and international guidelines including underlying literature was performed. RESULTS Nearly half of pancreatic cancer cases are missed by currently available surveillance programs, even in high-risk cohorts. Magnetic resonance imaging and endoscopic ultrasound supplemented by CA19‑9 (± HbA1c) are not accurate enough to ensure robust earlier pancreatic cancer detection. Complementary biomarker panels will take on a crucial diagnostic role in the future.
Collapse
Affiliation(s)
- Simon Sirtl
- Medizinische Klinik und Poliklinik II, LMU Klinikum, 81377, München, Deutschland.
| | - Marlies Vornhülz
- Medizinische Klinik und Poliklinik II, LMU Klinikum, 81377, München, Deutschland
| | - Felix O Hofmann
- Klinik für Allgemein‑, Viszeral- und Transplantationschirurgie, LMU Klinikum, München, Deutschland
| | - Julia Mayerle
- Medizinische Klinik und Poliklinik II, LMU Klinikum, 81377, München, Deutschland.
| | - Georg Beyer
- Medizinische Klinik und Poliklinik II, LMU Klinikum, 81377, München, Deutschland
| |
Collapse
|
46
|
Wang L, Grimshaw AA, Mezzacappa C, Larki NR, Yang YX, Justice AC. Do Polygenic Risk Scores Add to Clinical Data in Predicting Pancreatic Cancer? A Scoping Review. Cancer Epidemiol Biomarkers Prev 2023; 32:1490-1497. [PMID: 37610426 PMCID: PMC10873036 DOI: 10.1158/1055-9965.epi-23-0468] [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: 05/02/2023] [Revised: 07/21/2023] [Accepted: 08/21/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Polygenic risk scores (PRS) summarize an individual's germline genetic risk, but it is unclear whether PRS offer independent information for pancreatic cancer risk prediction beyond routine clinical data. METHODS We searched 8 databases from database inception to March 10, 2023 to identify studies evaluating the independent performance of pancreatic cancer-specific PRS for pancreatic cancer beyond clinical risk factors. RESULTS Twenty-one studies examined associations between a pancreatic cancer-specific PRS and pancreatic cancer. Seven studies evaluated risk factors beyond age and sex. Three studies evaluated the change in discrimination associated with the addition of PRS to routine risk factors and reported improvements (AUCs: 0.715 to 0.745; AUC 0.791 to 0.830; AUC from 0.694 to 0.711). Limitations to clinical applicability included using source populations younger/healthier than those at risk for pancreatic cancer (n = 10), exclusively of European ancestry (n = 13), or controls without relevant exposures (n = 1). CONCLUSIONS While most studies of pancreatic cancer-specific PRS did not evaluate the independent discrimination of PRS for pancreatic cancer beyond routine risk factors, three that did showed improvements in discrimination. IMPACT For pancreatic cancer PRS to be clinically useful, they must demonstrate substantial improvements in discrimination beyond established risk factors, apply to diverse ancestral populations representative of those at risk for pancreatic cancer, and use appropriate controls.
Collapse
Affiliation(s)
- Louise Wang
- VA Connecticut Healthcare System, West Haven, CT, USA
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Catherine Mezzacappa
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Navid Rahimi Larki
- VA Connecticut Healthcare System, West Haven, CT, USA
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Yu-Xiao Yang
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA USA
| | - Amy C. Justice
- VA Connecticut Healthcare System, West Haven, CT, USA
- Section of General Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- School of Public Health, Yale University, New Haven, CT, USA
| |
Collapse
|
47
|
Vitali F, Zundler S, Jesper D, Strobel D, Wildner D, de Pretis N, Frulloni L, Crinó SF, Neurath MF. Endoscopic Ultrasound in Pancreatology: Focus on Inflammatory Diseases and Interventions. Visc Med 2023; 39:131-139. [PMID: 37899796 PMCID: PMC10601533 DOI: 10.1159/000533433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/03/2023] [Indexed: 10/31/2023] Open
Abstract
Background Endoscopic ultrasound (EUS) is a main tool in pancreatology for both diagnosis and therapy. It allows minimally invasive differentiation of various diseases, with a minimal degree of inflammation or anatomic variations. EUS also enables interventional direct access to the pancreatic parenchyma and the retroperitoneal space, the pancreatic duct, the pancreatic masses, cysts, vascular structures for diagnostic and therapeutic purposes. Summary This review aimed to summarize the new developments of EUS in the field of pancreatology, with special interest on inflammation and interventions. EUS enables way to perform pseudocyst drainage, necrosectomy, transenteral drainage and transenteric access of the main pancreatic duct, or the direct visualization or therapy of vascular structures adjacent to the pancreas. Key Messages EUS has a deep impact on pancreatology, and the development of new diagnostic and interventional approaches to the retroperitoneal space and the pancreas has increased in the last years exponentially, allowing minimal invasive diagnostics and therapy and avoiding surgery and percutaneous therapy.
Collapse
Affiliation(s)
- Francesco Vitali
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Daniel Jesper
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Deike Strobel
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Dane Wildner
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Nicoló de Pretis
- Gastroenterology and Digestive Endoscopy Unit, The Pancreas Institute, G.B. Rossi University Hospital, Verona, Italy
| | - Luca Frulloni
- Gastroenterology and Digestive Endoscopy Unit, The Pancreas Institute, G.B. Rossi University Hospital, Verona, Italy
| | - Stefano Francesco Crinó
- Gastroenterology and Digestive Endoscopy Unit, The Pancreas Institute, G.B. Rossi University Hospital, Verona, Italy
| | - Markus F. Neurath
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
48
|
Lin W, Fang J, Wei S, He G, Liu J, Li X, Peng X, Li D, Yang S, Li X, Yang L, Li H. Extracellular vesicle-cell adhesion molecules in tumours: biofunctions and clinical applications. Cell Commun Signal 2023; 21:246. [PMID: 37735659 PMCID: PMC10512615 DOI: 10.1186/s12964-023-01236-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/18/2023] [Indexed: 09/23/2023] Open
Abstract
Cell adhesion molecule (CAM) is an umbrella term for several families of molecules, including the cadherin family, integrin family, selectin family, immunoglobulin superfamily, and some currently unclassified adhesion molecules. Extracellular vesicles (EVs) are important information mediators in cell-to-cell communication. Recent evidence has confirmed that CAMs transported by EVs interact with recipient cells to influence EV distribution in vivo and regulate multiple cellular processes. This review focuses on the loading of CAMs onto EVs, the roles of CAMs in regulating EV distribution, and the known and possible mechanisms of these actions. Moreover, herein, we summarize the impacts of CAMs transported by EVs to the tumour microenvironment (TME) on the malignant behaviour of tumour cells (proliferation, metastasis, immune escape, and so on). In addition, from the standpoint of clinical applications, the significance and challenges of using of EV-CAMs in the diagnosis and therapy of tumours are discussed. Finally, considering recent advances in the understanding of EV-CAMs, we outline significant challenges in this field that require urgent attention to advance research and promote the clinical applications of EV-CAMs. Video Abstract.
Collapse
Affiliation(s)
- Weikai Lin
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Jianjun Fang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Shibo Wei
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Guangpeng He
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Jiaxing Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Xian Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Dai Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Shuo Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China.
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China.
| |
Collapse
|
49
|
Karstensen JG, Bülow S, Højen H, Jelsig AM, Jespersen N, Andersen KK, Wewer MD, Burisch J, Pommergaard HC. Cancer in Patients With Familial Adenomatous Polyposis: A Nationwide Danish Cohort Study With Matched Controls. Gastroenterology 2023; 165:573-581.e3. [PMID: 37201686 DOI: 10.1053/j.gastro.2023.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/25/2023] [Accepted: 05/02/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND & AIMS Familial adenomatous polyposis (FAP) is a hereditary disorder that predisposes patients to colorectal cancer (CRC). Prophylactic colectomy has greatly reduced the risk of CRC. However, new associations between FAP and the risk of other cancers have subsequently emerged. In this study, we assessed the risk of specific primary and secondary cancers among patients with FAP compared with matched controls. METHODS All known patients with FAP up until April 2021 were identified in the nationwide Danish Polyposis Register and paired with 4 unique controls matched by birth year, sex, and postal code. The risk of overall cancers, specific cancer types, and risk of a second primary cancer was assessed and compared with controls. RESULTS The analysis included 565 patients with FAP and 1890 controls. The overall risk of cancer was significantly higher for patients with FAP than for controls (hazard ratio [HR], 4.12; 95% confidence interval [CI], 3.28-5.17; P < .001). The increased risk was mainly due to CRC (HR, 4.61; 95% CI, 2.58-8.22; P < .001), pancreatic cancer (HR, 6.45; 95% CI, 2.02-20.64; P = .002), and duodenal/small-bowel cancer (HR, 14.49; 95% CI, 1.76-119.47; P = .013), whereas no significant difference was observed for gastric cancer (HR, 3.29; 95% CI, 0.53-20.23; P = .20). Furthermore, the risk of a second primary cancer was significantly higher for patients with FAP (HR, 1.89; 95% CI, 1.02-3.50; P = .042). Between 1980 and 2020, the risk of cancer among patients with FAP decreased by ∼50%. CONCLUSIONS Despite an absolute reduction in the risk of developing cancer among patients with FAP, the risk remained significantly higher than for the background population due to colorectal, pancreatic, and duodenal/small-bowel cancers.
Collapse
Affiliation(s)
- John Gásdal Karstensen
- Danish Polyposis Register, Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | - Steffen Bülow
- Danish Polyposis Register, Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | - Helle Højen
- Danish Polyposis Register, Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | - Anne Marie Jelsig
- Department of Clinical Genetics, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Niels Jespersen
- Danish Polyposis Register, Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | | | - Mads Damsgaard Wewer
- Danish Polyposis Register, Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark; Gastro Unit, Medical Division, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | - Johan Burisch
- Danish Polyposis Register, Gastro Unit, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark; Gastro Unit, Medical Division, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | - Hans Christian Pommergaard
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Department of Surgery and Transplantation, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
50
|
Sijithra PC, Santhi N, Ramasamy N. A review study on early detection of pancreatic ductal adenocarcinoma using artificial intelligence assisted diagnostic methods. Eur J Radiol 2023; 166:110972. [PMID: 37454557 DOI: 10.1016/j.ejrad.2023.110972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive, chemo-refractory and recalcitrant cancer and increases the number of deaths. With just around 1 in 4 individuals having respectable tumours, PDAC is frequently discovered when it is in an advanced stage. Accordingly, ED of PDAC improves patient survival. Subsequently, this paper reviews the early detection of PDAC, initially, the work presented an overview of PDAC. Subsequently, it reviews the molecular biology of pancreatic cancer and the development of molecular biomarkers are represented. This article illustrates the importance of identifying PDCA, the Immune Microenvironment of Pancreatic Cancer. Consequently, in this review, traditional and non-traditional imaging techniques are elucidated, traditional and non-traditional methods like endoscopic ultrasound, Multidetector CT, CT texture analysis, PET-CT, magnetic resonance imaging, diffusion-weighted imaging, secondary signs of pancreatic cancer, and molecular imaging. The use of artificial intelligence in pancreatic cancer, novel MRI techniques, and the future directions of AI for PDAC detection and prognosis is then described. Additionally, the research problem definition and motivation, current trends and developments, state of art of survey, and objective of the research are demonstrated in the review. Consequently, this review concluded that Artificial Intelligence Assisted Diagnostic Methods with MRI images can be proposed in future to improve the specificity and the sensitivity of the work, and to classify malignant PDAC with greater accuracy.
Collapse
Affiliation(s)
- P C Sijithra
- Department of Electronics and Communication Engineering, Noorul Islam Centre for Higher Education, Kanyakumari District, Tamilnadu, India.
| | - N Santhi
- Department of Electronics and Communication Engineering, Noorul Islam Centre for Higher Education, Kanyakumari District, Tamilnadu, India
| | - N Ramasamy
- Department of Mechanical Engineering, Noorul Islam Centre for Higher Education, Kanyakumari District, Tamilnadu, India
| |
Collapse
|