1
|
Farinella R, Felici A, Peduzzi G, Testoni SGG, Costello E, Aretini P, Blazquez-Encinas R, Oz E, Pastore A, Tacelli M, Otlu B, Campa D, Gentiluomo M. From classical approaches to artificial intelligence, old and new tools for PDAC risk stratification and prediction. Semin Cancer Biol 2025; 112:71-92. [PMID: 40147701 DOI: 10.1016/j.semcancer.2025.03.004] [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: 11/28/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is recognized as one of the most lethal malignancies, characterized by late-stage diagnosis and limited therapeutic options. Risk stratification has traditionally been performed using epidemiological studies and genetic analyses, through which key risk factors, including smoking, diabetes, chronic pancreatitis, and inherited predispositions, have been identified. However, the multifactorial nature of PDAC has often been insufficiently addressed by these methods, leading to limited precision in individualized risk assessments. Advances in artificial intelligence (AI) have been proposed as a transformative approach, allowing the integration of diverse datasets-spanning genetic, clinical, lifestyle, and imaging data into dynamic models capable of uncovering novel interactions and risk profiles. In this review, the evolution of PDAC risk stratification is explored, with classical epidemiological frameworks compared to AI-driven methodologies. Genetic insights, including genome-wide association studies and polygenic risk scores, are discussed, alongside AI models such as machine learning, radiomics, and deep learning. Strengths and limitations of these approaches are evaluated, with challenges in clinical translation, such as data scarcity, model interpretability, and external validation, addressed. Finally, future directions are proposed for combining classical and AI-driven methodologies to develop scalable, personalized predictive tools for PDAC, with the goal of improving early detection and patient outcomes.
Collapse
Affiliation(s)
| | | | | | - Sabrina Gloria Giulia Testoni
- Division of Gastroenterology and Gastrointestinal Endoscopy, IRCCS Policlinico San Donato, Vita-Salute San Raffaele University, Milan, Italy
| | - Eithne Costello
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Paolo Aretini
- Fondazione Pisana per la Scienza, San Giuliano Terme, Italy
| | - Ricardo Blazquez-Encinas
- Department of Cell Biology, Physiology and Immunology, University of Cordoba / Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Elif Oz
- Department of Biostatistics and Bioinformatics, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Aldo Pastore
- Fondazione Pisana per la Scienza, San Giuliano Terme, Italy
| | - Matteo Tacelli
- Pancreas Translational & Clinical Research Center, Pancreato-Biliary Endoscopy and Endosonography Division, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Burçak Otlu
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, Ankara, Turkey
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | | |
Collapse
|
2
|
Murray K, Oldfield L, Stefanova I, Gentiluomo M, Aretini P, O'Sullivan R, Greenhalf W, Paiella S, Aoki MN, Pastore A, Birch-Ford J, Rao BH, Uysal-Onganer P, Walsh CM, Hanna GB, Narang J, Sharma P, Campa D, Rizzato C, Turtoi A, Sever EA, Felici A, Sucularli C, Peduzzi G, Öz E, Sezerman OU, Van der Meer R, Thompson N, Costello E. Biomarkers, omics and artificial intelligence for early detection of pancreatic cancer. Semin Cancer Biol 2025; 111:76-88. [PMID: 39986585 DOI: 10.1016/j.semcancer.2025.02.009] [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: 11/29/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is frequently diagnosed in its late stages when treatment options are limited. Unlike other common cancers, there are no population-wide screening programmes for PDAC. Thus, early disease detection, although urgently needed, remains elusive. Individuals in certain high-risk groups are, however, offered screening or surveillance. Here we explore advances in understanding high-risk groups for PDAC and efforts to implement biomarker-driven detection of PDAC in these groups. We review current approaches to early detection biomarker development and the use of artificial intelligence as applied to electronic health records (EHRs) and social media. Finally, we address the cost-effectiveness of applying biomarker strategies for early detection of PDAC.
Collapse
Affiliation(s)
- Kate Murray
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Lucy Oldfield
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Irena Stefanova
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | | | | | - Rachel O'Sullivan
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - William Greenhalf
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Salvatore Paiella
- Pancreatic Surgery Unit, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Italy
| | - Mateus N Aoki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Brazil
| | - Aldo Pastore
- Fondazione Pisana per la Scienza, Scuola Normale Superiore di Pisa, Italy
| | - James Birch-Ford
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Bhavana Hemantha Rao
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Czech Republic
| | - Pinar Uysal-Onganer
- School of Life Sciences, Cancer Mechanisms and Biomarkers Group, The University of Westminster, United Kingdom
| | - Caoimhe M Walsh
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - George B Hanna
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | | | | | | | | | - Andrei Turtoi
- Tumor Microenvironment and Resistance to Treatment Lab, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, France
| | - Elif Arik Sever
- Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | | | | | | | - Elif Öz
- Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | - Osman Uğur Sezerman
- Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | | | | | - Eithne Costello
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom.
| |
Collapse
|
3
|
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
|
4
|
Liu PJ, Zhou ZP, Wang GY, Xu S, Wang W, Chen X, Tan XD, Liu ZH, Zhao ZM, Gao YX, Zhang XP, Liu R. New-onset diabetes worsens prognosis of patients with pancreatic ductal adenocarcinoma after R0 resection: A multicenter study. Hepatobiliary Pancreat Dis Int 2025:S1499-3872(25)00088-8. [PMID: 40374469 DOI: 10.1016/j.hbpd.2025.04.008] [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: 11/11/2024] [Accepted: 04/03/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND The risk of pancreatic ductal adenocarcinoma (PDAC) is increased in patients with diabetes mellitus (DM), particularly in new-onset diabetes (NOD). This study aimed to analyze the effect of NOD on the outcomes of patients with PDAC after R0 resection. METHODS PDAC patients from six centers in China undergoing R0 resection from 2015 to 2022 were included. Patients were categorized as long-term diabetes (LTD), NOD, or non-diabetes mellitus (non-DM) based on the timing of diagnosis relative to pancreatic resection. We compared the effects of diabetes status on perioperative and oncological outcomes of PDAC. RESULTS Of 1211 patients, 602 (49.7%), 127 (10.5%), and 482 (39.8%) were in the non-DM, LTD, and NOD groups, respectively. Patients with NOD suffered from higher rates of fatty pancreas and postoperative pancreatic fistula (POPF) (both P < 0.05). When compared with the non-DM group, the NOD group had worse median overall survival (OS) (24.6 vs. 29.4 months, P < 0.001) and recurrence-free survival (RFS) (13.3 vs. 15.8 months, P < 0.001); and the LTD group also had worse median OS (25.2 vs. 29.4 months, P = 0.041) and RFS (13.8 vs. 15.8 months, P = 0.007) compared with non-DM group. However, there were no significant differences in survival between the NOD and the LTD groups. Multivariate analysis indicated that NOD, LTD, largest tumor size, and poor tumor differentiation were independently associated with worse OS and RFS (all P < 0.05). CONCLUSIONS Patients with PDAC undergoing R0 resection experienced a higher probability of POPF in the presence of concurrent NOD. Long-term survival prognosis was worse in NOD or LTD patients than in non-DM patients.
Collapse
Affiliation(s)
- Peng-Jiong Liu
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Zhi-Peng Zhou
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Guan-Yu Wang
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Shuai Xu
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Wei Wang
- Department of General Surgery, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Xiong Chen
- Department of Hepatobiliary Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, China
| | - Xiao-Dong Tan
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Zhong-Hua Liu
- Department of Hepatobiliary Surgery, Chifeng Municipal Hospital, Chifeng 024050, China
| | - Zhi-Ming Zhao
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Yuan-Xing Gao
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Xiu-Ping Zhang
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| | - Rong Liu
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
5
|
Munigala S, Bowe B, Subramaniam DS, Xian H, Gowda AN, Sheth SG, Chhabra R, Burroughs TE, Agarwal B. Assessing NODM Patients for Early PDAC Diagnosis: Incidence of NODM Before PDAC Diagnosis and Subsequent PDAC Risk. Cancer Med 2025; 14:e70878. [PMID: 40351037 PMCID: PMC12066942 DOI: 10.1002/cam4.70878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/11/2025] [Accepted: 04/04/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND New-Onset Diabetes Mellitus (NODM) is often an early manifestation of pancreatic cancer (Pancreatic Ductal Adenocarcinoma, PDAC). However, there is limited information about (1) the duration prior to PDAC diagnosis when the annual incidence of NODM starts significantly exceeding that in age-matched controls, (2) the percentage of PDAC patients diagnosed with NODM in the years preceding, and (3) the risk of PDAC following NODM in time when the PDAC risk is significantly higher than in controls. METHODS Using the nationwide VA database, we evaluated the annual incidence of NODM for 15 years preceding the PDAC diagnosis and in the age- and sex-matched controls (1:5 matching). In the second part, we evaluated the long-term risk and predictors of PDAC in NODM patients and controls. RESULTS The case-control study comprised 8198 PDAC patients and 40,992 matched controls. The higher annual incidence of NODM in PDAC patients was statistically significant up to 15 years before PDAC diagnosis. 69.2% of PDAC patients had NODM in the preceding 15 years versus 38.0% of controls. PDAC risk in the 15 years following NODM was 0.60% compared to 0.13% in the controls (aHR 3.83, 95% CI 3.68-3.98, p < 0.001). The risk of PDAC is more pronounced in the 1 year following NODM (aHR 9.07, 95% CI 8.33-9.87) than the subsequent 5 years (aHR 2.98, 95% CI 2.82-3.15). CONCLUSION NODM pre-dates PDAC diagnosis in most patients with PDAC. Further evaluation of NODM patients for PDAC has the potential to become a feasible strategy for diagnosing more early-stage resectable PDACs.
Collapse
Affiliation(s)
- Satish Munigala
- College for Public Health and Social JusticeSaint Louis UniversitySaint LouisMissouriUSA
- Department of Internal MedicineWashington University in St. LouisSaint LouisMissouriUSA
| | - Benjamin Bowe
- Clinical Epidemiology Center, Research and Education Service, VA Saint Louis, Health Care SystemSaint LouisMissouriUSA
| | - Divya S. Subramaniam
- Department of Health and Clinical Outcomes ResearchSaint Louis University School of MedicineSaint LouisMissouriUSA
- Advanced HEAlth Data (AHEAD) InstituteSaint Louis UniversitySaint LouisMissouriUSA
| | - Hong Xian
- Department of Epidemiology & Biostatistics, College for Public Health and Social JusticeSaint Louis UniversitySaint LouisMissouriUSA
| | | | - Sunil G. Sheth
- Beth Israel Deaconess Medical Center, and Harvard Medical SchoolBostonMassachusettsUSA
| | - Rajiv Chhabra
- Saint Luke's Hospital and University of Missouri Kansas CityKansas CityMissouriUSA
| | - Thomas E. Burroughs
- College for Public Health and Social JusticeSaint Louis UniversitySaint LouisMissouriUSA
- Department of Internal MedicineWashington University in St. LouisSaint LouisMissouriUSA
| | - Banke Agarwal
- Department of GastroenterologySSM St. Anthony's HospitalOklahoma CityOklahomaUSA
| |
Collapse
|
6
|
Zhou Y, Wu Z, Zeng L, Chen R. Combining genetic and non-genetic factors to predict the risk of pancreatic cancer in patients with new-onset diabetes mellitus. BMC Med 2025; 23:224. [PMID: 40234846 PMCID: PMC12001390 DOI: 10.1186/s12916-025-04048-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 04/02/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Recent research suggests that new-onset diabetes mellitus (NODM) often results from pancreatic cancer (PC) rather than causing it. Determining if NODM is type 2 diabetes mellitus (T2DM) or PC-related NODM (NODM-PC) aids in the early diagnosis of PC. We developed a NODM-PC risk prediction model to stratify PC risk in patients with NODM. METHODS This study utilized data from the UK Biobank, including 238 NODM-PC cases and 14,825 cancer-free T2DM controls. Polygenic risk scores (PRSs) for PC and T2DM were constructed using previously reported single nucleotide polymorphisms (SNPs) separately, while the NODM-PC PRS was developed by combining SNPs from both. Non-genetic factors were selected as candidate predictors based on prior NODM-PC prediction models. We developed three Cox models to estimate the risk of PC diagnosis within 3 years in the NODM population and evaluated them by internal-external cross-validation. RESULTS Elevated NODM-PC PRS and PC PRS scores positively correlated with NODM-PC risk, while T2DM PRS showed an inverse correlation. The NODM-PC PRS achieved the highest AUC at 0.719. Three Cox models were developed: Model 1 included age at T2DM diagnosis, smoking status, HbA1c, PC PRS, and T2DM PRS; Model 2 replaced PC and T2DM PRS with NODM-PC PRS; Model 3 included only non-genetic factors. Model 2 had the highest discrimination (Harrell's C-index 0.823 (95% CI: 0.806-0.840)), demonstrated the best clinical utility with good calibration, and showed significant classification improvement (continuous net reclassification index: 26.89% and 31.93% for cases, 28.51% and 30.90% for controls, compared to Models 1 and 3). The positive predictive value for the top 1% predicted risk in Model 2 was 13.25%. CONCLUSIONS This NODM-PC PRS enhances NODM-PC risk prediction, efficiently identifies individuals at high risk for PC screening, and improves PC screening efficiency at the population level among NODM individuals.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhuo Wu
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Liangtang Zeng
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China.
| | - Rufu Chen
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China.
| |
Collapse
|
7
|
Lewis D, Jiménez L, Chan KK, Horton S, Wong WWL. A Systematic Review of Cost-Effectiveness Studies on Pancreatic Cancer Screening. Curr Oncol 2025; 32:225. [PMID: 40277782 PMCID: PMC12025814 DOI: 10.3390/curroncol32040225] [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: 02/26/2025] [Revised: 03/29/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is among the deadliest types of cancer globally. While early detection helps avert adverse outcomes, screening is only recommended for individuals at high risk, specifically those with familial and/or genetic predispositions. The objectives of this study are to systematically review primary studies on the cost-effectiveness of PC screening and to identify the critical factors that influence cost-effectiveness. METHODS This systematic review was performed using PRISMA guidelines. Economic evaluation studies on PC screening were identified from searches on the SCOPUS and PubMed databases. The quality of reporting of the selected articles was assessed according to CHEERS 2022. Using predefined inclusion and exclusion criteria, two reviewers conducted the title-abstract review, full-text review, and data extraction to select relevant articles. The authors' consensus was used to settle disagreements. The primary outcome was the incremental cost-effectiveness ratio, measured by cost per quality-adjusted life year and cost per life year saved. RESULTS Nine studies were selected for the final review. Most studies demonstrated that one-time screening for PC among high-risk individuals was cost-effective compared with no screening, while others found annual screening to also be cost-effective. High-risk was generally defined as having a >5% lifetime risk of PC and included individuals with either familial pancreatic cancer (FPC) or genetic susceptibility syndromes such as Peutz-Jeghers Syndrome, hereditary pancreatitis, hereditary non-polypoid colorectal cancer syndrome, familial adenomatous polyposis, and BRCA2 mutations. Individuals with new-onset diabetes (NOD) were also considered high-risk. Screening using mainly endoscopic ultrasound was cost-effective among FPC individuals and those with genetic syndromes. Risk-based screening was also cost-effective among patients with NOD. CONCLUSION Screening for PC is cost-effective among selected high-risk individuals. However, cost-effectiveness depends on epidemiological factors, cost, the diagnostic performance of screening tools, and the overall design of studies.
Collapse
Affiliation(s)
- Diedron Lewis
- School of Pharmacy, University of Waterloo, Waterloo, ON N2G 1C5, Canada;
| | - Laura Jiménez
- Department of Community Health and Epidemiology, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Kelvin K. Chan
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
| | - Susan Horton
- School of Public Health Sciences, University of Waterloo, Waterloo, ON N2L 3G5, Canada;
| | - William W. L. Wong
- School of Pharmacy, University of Waterloo, Waterloo, ON N2G 1C5, Canada;
| |
Collapse
|
8
|
Zhu W, Chen L, Aphinyanaphongs Y, Kastrinos F, Simeone DM, Pochapin M, Stender C, Razavian N, Gonda TA. Identification of patients at risk for pancreatic cancer in a 3-year timeframe based on machine learning algorithms. Sci Rep 2025; 15:11697. [PMID: 40188106 PMCID: PMC11972345 DOI: 10.1038/s41598-025-89607-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 02/06/2025] [Indexed: 04/07/2025] Open
Abstract
Early detection of pancreatic cancer (PC) remains challenging largely due to the low population incidence and few known risk factors. However, screening in at-risk populations and detection of early cancer has the potential to significantly alter survival. In this study, we aim to develop a predictive model to identify patients at risk for developing new-onset PC at two and a half to three year time frame. We used the Electronic Health Records (EHR) of a large medical system from 2000 to 2021 (N = 537,410). The EHR data analyzed in this work consists of patients' demographic information, diagnosis records, and lab values, which are used to identify patients who were diagnosed with pancreatic cancer and the risk factors used in the machine learning algorithm for prediction. We identified 73 risk factors of pancreatic cancer with the Phenome-wide Association Study (PheWAS) on a matched case-control cohort. Based on them, we built a large-scale machine learning algorithm based on EHR. A temporally stratified validation based on patients not included in any stage of the training of the model was performed. This model showed an AUROC at 0.742 [0.727, 0.757] which was similar in both the general population and in a subset of the population who has had prior cross-sectional imaging. The rate of diagnosis of pancreatic cancer in those in the top 1 percentile of the risk score was 6 folds higher than the general population. Our model leverages data extracted from a 6-month window of time in the electronic health record to identify patients at nearly sixfold higher than baseline risk of developing pancreatic cancer 2.5-3 years from evaluation. This approach offers an opportunity to define an enriched population entirely based on static data, where current screening may be recommended.
Collapse
Affiliation(s)
- Weicheng Zhu
- Center for Data Science, New York University, New York, NY, USA
| | - Long Chen
- Center for Data Science, New York University, New York, NY, USA
| | - Yindalon Aphinyanaphongs
- Department of Population Health, New York University Grossman School of Medicine, 227 East 30th Street, 6th Floor, New York, NY, 10016, USA
| | - Fay Kastrinos
- Department of Medicine, Division of Digestive and Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Diane M Simeone
- Moores Cancer Center, UC San Diego Health, San Diego, CA, USA
| | - Mark Pochapin
- Division of Gastroenterology and Hepatology, Department of Medicine, New York University, 240 East 38th Street, 23rd Floor, New York, NY, 10016, USA
| | - Cody Stender
- Department of Surgery, New York University, New York, NY, USA
| | - Narges Razavian
- Department of Population Health, New York University Grossman School of Medicine, 227 East 30th Street, 6th Floor, New York, NY, 10016, USA.
| | - Tamas A Gonda
- Division of Gastroenterology and Hepatology, Department of Medicine, New York University, 240 East 38th Street, 23rd Floor, New York, NY, 10016, USA.
| |
Collapse
|
9
|
Kazerouni AR, Ghahramani S, Khayyer Y, Yousufzai S. New-onset type 2 diabetes mellitus complicated by diabetic ketoacidosis: a sentinel presentation of advanced pancreatic adenocarcinoma. Endocrinol Diabetes Metab Case Rep 2025; 2025:e250026. [PMID: 40421653 PMCID: PMC12122053 DOI: 10.1530/edm-25-0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 05/02/2025] [Accepted: 05/16/2025] [Indexed: 05/28/2025] Open
Abstract
Summary Diabetic ketoacidosis (DKA), typically linked to type 1 diabetes or acute illness in type 2 diabetes, can rarely be triggered by pancreatic adenocarcinoma (PA). Though 80% of PA patients have glucose intolerance, DKA is exceptionally uncommon, with fewer than 20 documented cases. A 52-year-old woman with new-onset type 2 diabetes presented with altered mental status, abdominal pain, and 23 kg weight loss over 2 months. Labs confirmed DKA (glucose: 439 mg/dL, pH 7.1, ketonuria). Elevated tumor markers (CA19-9: >10,000 U/mL, CEA: 365 ng/mL) and imaging revealed a 4 cm pancreatic mass with metastases, biopsy-proven as PA. This case underscores PA as a rare but critical DKA precipitant in new-onset diabetes. Unexplained hyperglycemia, rapid weight loss, and markedly elevated tumor markers should prompt malignancy screening. Early multidisciplinary intervention may improve outcomes in this aggressive cancer. Clinicians must maintain high suspicion for occult PA in atypical DKA presentations. Learning points Unexplained weight loss alongside newly-identified type 2 DM warrants thorough evaluation for occult malignancy. Elevated CA19-9 and CEA in the context of new-onset diabetes should raise suspicion for pancreatic malignancy. DKA may rarely serve as the initial manifestation of pancreatic cancer in newly-identified type 2 DM cases, necessitating a high index of clinical suspicion.
Collapse
Affiliation(s)
- Akbar Rasekhi Kazerouni
- Department of Internal Medicine, Gastroenterohepatology Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Sahar Ghahramani
- Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Khayyer
- Department of Pathology, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Shayan Yousufzai
- Student Committee of Medical Education Development, Education Development Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
10
|
Wang Y, Kim MP. Guidelines for the management of pancreatic cystic lesions: many options, too few solutions? Hepatobiliary Surg Nutr 2025; 14:345-347. [PMID: 40342772 PMCID: PMC12057492 DOI: 10.21037/hbsn-2025-108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 05/11/2025]
Affiliation(s)
- Yifan Wang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael P. Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
11
|
Mejza M, Bajer A, Wanibuchi S, Małecka-Wojciesko E. Can AI Be Useful in the Early Detection of Pancreatic Cancer in Patients with New-Onset Diabetes? Biomedicines 2025; 13:836. [PMID: 40299428 PMCID: PMC12025102 DOI: 10.3390/biomedicines13040836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/12/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Pancreatic cancer is one of the most lethal neoplasms. Despite considerable research conducted in recent decades, not much has been achieved to improve its survival rate. That may stem from the lack of effective screening strategies in increased pancreatic cancer risk groups. One population that may be appropriate for screening is new-onset diabetes (NOD) patients. Such a conclusion stems from the fact that pancreatic cancer can cause diabetes several months before diagnosis. The most widely used screening tool for this population, the ENDPAC (Enriching New-Onset Diabetes for Pancreatic Cancer) model, has not achieved satisfactory results in validation trials. This provoked the first attempts at using artificial intelligence (AI) to create larger, multi-parameter models that could better identify the at-risk population, which would be suitable for screening. The results shown by the authors of these trials seem promising. Nonetheless, the number of publications is limited, and the downfalls of using AI are not well highlighted. This narrative review presents a summary of previous publications, recent advancements and feasible solutions for effective screening of patients with NOD for pancreatic cancer.
Collapse
Affiliation(s)
- Maja Mejza
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-153 Lodz, Poland; (M.M.); (A.B.)
| | - Anna Bajer
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-153 Lodz, Poland; (M.M.); (A.B.)
| | - Sora Wanibuchi
- Aichi Medical University Hospital, Nagakute 480-1195, Japan;
| | - Ewa Małecka-Wojciesko
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-153 Lodz, Poland; (M.M.); (A.B.)
| |
Collapse
|
12
|
Tan JT, Mao X, Cheng HM, Seto WK, Leung WK, Cheung KS. Aspirin is associated with lower risk of pancreatic cancer and cancer-related mortality in patients with diabetes mellitus. Gut 2025; 74:603-612. [PMID: 39746785 DOI: 10.1136/gutjnl-2024-333329] [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: 07/11/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Patients with type 2 diabetes mellitus (T2DM) have higher pancreatic cancer (PC) risk. While aspirin has chemopreventive effects on digestive cancers, its effect on PC among patients with T2DM is unclear. METHODS This retrospective cohort study identified newly diagnosed adult patients with T2DM in Hong Kong between 2001 and 2015 from a territory-wide healthcare registry. Exclusion criteria were history of PC, pancreatic cyst, IgG4 disease, or pancreatectomy. To address reverse causality between PC and T2DM, we excluded patients with PC diagnosed within 1 year of T2DM. We also excluded patients with less than 1 year of observation. Primary outcome was PC, and secondary outcomes were PC-related and all-cause mortality. Aspirin use was treated as time-varying variable (≥180 day-use/year) to address immortal-time bias, and multivariable Cox regression model was employed to derive adjusted HR (aHR). Propensity-score (PS) matching was used as secondary analysis. RESULTS Among 343 966 newly diagnosed patients with T2DM (median follow-up: 10.5 years; IQR: 7.7-14.5 years), 1224 (0.36%) developed PC. There were 51 151 (14.9%) deaths from any cause, and 787 (0.2%) died from PC. Aspirin use was associated with lower PC risk in both time-dependent (aHR: 0.58; 95% CI 0.49 to 0.69) and PS matching analysis (aHR: 0.61; 95% CI 0.48 to 0.77). An inverse relationship was observed with increasing dose and duration of aspirin use (P trend<0.001). Aspirin was also associated with a lower risk of PC-related mortality (aHR: 0.43; 95% CI 0.34 to 0.53) and all-cause mortality (aHR: 0.78; 95% CI 0.76 to 0.80). CONCLUSION Aspirin use may be an oncopreventive strategy to reduce PC risk in patients with T2DM. Further studies are warranted to validate the study findings.
Collapse
Affiliation(s)
- Jing Tong Tan
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Xianhua Mao
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ho-Ming Cheng
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Wai-Kay Seto
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Wai-K Leung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Ka-Shing Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
13
|
Rodriguez J, Coté GA. Clinical and Investigative Approach to Recurrent Acute Pancreatitis. Gastroenterol Clin North Am 2025; 54:113-127. [PMID: 39880522 DOI: 10.1016/j.gtc.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Recurrent acute pancreatitis (RAP) is a complex syndrome that presents variably, with many cases remaining idiopathic after thorough diagnostics. For evaluating structural etiologies, endoscopic ultrasound and MR cholangiopancreatography are preferred over endoscopic retrograde cholangiopancreatography (ERCP) given their more favorable risk profile and sensitivity. The diagnostic work-up remains paramount since treatment should focus on addressing underlying causes such as early cholecystectomy for gallstone pancreatitis. As more etiologic factors are uncovered, such as genetic susceptibility, causality becomes more nuanced. Earlier enthusiasm for endoscopic sphincterotomy as a treatment for idiopathic RAP has been tempered by less favorable studies in recent years.
Collapse
Affiliation(s)
- Jennifer Rodriguez
- Division of Gastroenterology and Hepatology, Oregon Health & Science University, Portland, OR, USA
| | - Gregory A Coté
- Division of Gastroenterology and Hepatology, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
14
|
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
|
15
|
Mohindroo C, Dy PS, Hande SP, D'Adamo CR, Mavanur A, Thomas A, McAllister F, De Jesus-Acosta A. New onset diabetes predicts clinical outcomes in patients with pancreatic adenocarcinoma. J Gastrointest Oncol 2025; 16:226-233. [PMID: 40115918 PMCID: PMC11921277 DOI: 10.21037/jgo-24-570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/12/2024] [Indexed: 03/23/2025] Open
Abstract
Background One percent of pancreatic adenocarcinoma (PDAC) patients are diagnosed with new onset diabetes (NOD) over the age of 50 years within 3 years. Therefore, NOD is a major factor for early diagnosis of PDAC. Research has focused on understanding the differences between NOD and type 2 diabetes, particularly in relation to PDAC. However, conflicting data exists regarding their impact on survival outcomes in PDAC patients. We performed this multi-center study to assess the prevalence and influence of NOD on clinical outcomes in patients with PDAC within a community-based hospital system. Methods We conducted a retrospective cohort study of 138 patients with biopsy-proven PDAC with localized/borderline disease (n=70), and metastatic disease (n=68) at three institutions from 2014 to 2021. NOD group consisted of pts diagnosed with diabetes [hemoglobin A1c (HbA1c) >6.5%] or pre-diabetes (HbA1c 5.7-6.4%) within the 3 years prior to PDAC diagnosis. Primary aim of the study was to determine the impact of NOD on clinical outcomes. Results A total of 138 patients were included in the study, from which 30 met the criteria for NOD. No significant differences were noted in the demographic and clinical characteristics comparing patients based on NOD history. Comparing survival outcomes, NOD group was associated with worse overall survival (OS) in both the metastatic cohort [n=68, progression-free survival (PFS) 4.6 vs. 7.1 months, P=0.07; OS 7.1 vs. 13.2 months, P=0.01) and the resected cohort (n=40, PFS 8.4 vs. 19.3 months, P=0.04; OS 24.5 vs. 42.3 months, P=0.04). In multivariate analysis, the impact of NOD remained significant for OS and PFS in the resected cohort. Identifying common features amongst the NOD group, we found the entire cohort had a significant reduction in individual body mass index (BMI) 1 year prior to the NOD diagnosis (P=0.006). Conclusions NOD is associated with worse survival outcomes in patients with metastatic and resected PDAC. Reduction of BMI prior to diagnosis of NOD, warrants further investigation to be incorporated into the PDAC screening paradigm.
Collapse
Affiliation(s)
- Chirayu Mohindroo
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Paul S Dy
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Suraj P Hande
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Christopher R D'Adamo
- Department of Family and Community Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Arun Mavanur
- Department of Surgery, Sinai Hospital of Baltimore, Baltimore, MD, USA
- Department of Surgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Asha Thomas
- Division of Endocrinology, Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Florencia McAllister
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ana De Jesus-Acosta
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
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
|
17
|
Tacelli M, Lauri G, Tabacelia D, Tieranu CG, Arcidiacono PG, Săftoiu A. Integrating artificial intelligence with endoscopic ultrasound in the early detection of bilio-pancreatic lesions: Current advances and future prospects. Best Pract Res Clin Gastroenterol 2025; 74:101975. [PMID: 40210329 DOI: 10.1016/j.bpg.2025.101975] [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: 11/29/2024] [Accepted: 12/31/2024] [Indexed: 04/12/2025]
Abstract
The integration of Artificial Intelligence (AI) in endoscopic ultrasound (EUS) represents a transformative advancement in the early detection and management of biliopancreatic lesions. This review highlights the current state of AI-enhanced EUS (AI-EUS) for diagnosing solid and cystic pancreatic lesions, as well as biliary diseases. AI-driven models, including machine learning (ML) and deep learning (DL), have shown significant improvements in diagnostic accuracy, particularly in distinguishing pancreatic ductal adenocarcinoma (PDAC) from benign conditions and in the characterization of pancreatic cystic neoplasms. Advanced algorithms, such as convolutional neural networks (CNNs), enable precise image analysis, real-time lesion classification, and integration with clinical and genomic data for personalized care. In biliary diseases, AI-assisted systems enhance bile duct visualization and streamline diagnostic workflows, minimizing operator dependency. Emerging applications, such as AI-guided EUS fine-needle aspiration (FNA) and biopsy (FNB), improve diagnostic yields while reducing errors. Despite these advancements, challenges remain, including data standardization, model interpretability, and ethical concerns regarding data privacy. Future developments aim to integrate multimodal imaging, real-time procedural support, and predictive analytics to further refine the diagnostic and therapeutic potential of AI-EUS. AI-driven innovation in EUS stands poised to revolutionize pancreatico-biliary diagnostics, facilitating earlier detection, enhancing precision, and paving the way for personalized medicine in gastrointestinal oncology and beyond.
Collapse
Affiliation(s)
- Matteo Tacelli
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy.
| | - Gaetano Lauri
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy; "Vita-Salute" San Raffaele University, Milan, Italy
| | - Daniela Tabacelia
- Department of Gastroenterology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Universitatea de Medicină și Farmacie Carol Davila din București, Bucuresti, Romania
| | - Cristian George Tieranu
- Department of Gastroenterology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Universitatea de Medicină și Farmacie Carol Davila din București, Bucuresti, Romania
| | - Paolo Giorgio Arcidiacono
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy; "Vita-Salute" San Raffaele University, Milan, Italy
| | - Adrian Săftoiu
- Department of Gastroenterology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Universitatea de Medicină și Farmacie Carol Davila din București, Bucuresti, Romania
| |
Collapse
|
18
|
Lauri G, Mills K, Majumder S, Capurso G. The exposome as a target for primary prevention and a tool for early detection of pancreatic cancer. Best Pract Res Clin Gastroenterol 2025; 74:101991. [PMID: 40210335 DOI: 10.1016/j.bpg.2025.101991] [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] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 04/12/2025]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a highly aggressive malignancy with limited survival due to late stage diagnosis and scarce therapeutic options. Emerging evidence highlights the role of the "exposome," which encompasses environmental, lifestyle, and metabolic exposures, as a crucial determinant in PDAC risk and a potential avenue for early detection. This review synthesizes findings on modifiable risk factors, including smoking, obesity, diabetes, diet, and alcohol consumption, and their interplay with genetic and metabolic profiles in PDAC development. Additionally, we explore cutting-edge approaches in exposomic research, such as biobanking, electronic health record analysis, and AI-driven predictive models, to identify early biomarkers and stratify high-risk populations. This integrated framework aims to inform prevention strategies and improve early detection of PDAC.
Collapse
Affiliation(s)
- Gaetano Lauri
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Krystal Mills
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Gabriele Capurso
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
19
|
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
|
20
|
Zhang W, Chen J, Zhang W, Xu M. Advances in Endoscopic Ultrasound in Pancreatic Cancer Screening, Diagnosis, and Palliative Care. Biomedicines 2024; 13:76. [PMID: 39857661 PMCID: PMC11762820 DOI: 10.3390/biomedicines13010076] [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: 12/09/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy with a profoundly poor prognosis. Clinically, the condition most frequently manifests with symptoms including painless jaundice, abdominal discomfort, and back pain. Early diagnosis and the implementation of effective therapeutic strategies are critical for improving patient survival outcomes. However, merely 10-20% of patients are diagnosed at an early stage, with the majority presenting at advanced stages, often with metastasis. Consequently, early detection and intervention are crucial for enhancing prognosis. The widespread adoption of endoscopic ultrasonography (EUS) technology in recent years has significantly enhanced the diagnostic accuracy for pancreatic space-occupying lesions. EUS is increasingly recognized for its pivotal role in alleviating malignant biliary obstruction (MBO), gastric outlet obstruction (GOO), and refractory pain in advanced pancreatic cancer. This article aims to provide an overall review of the current applications of EUS in the diagnosis and treatment of pancreatic cancer, exploring its advantages and limitations in early screening, diagnosis, and palliative care. Furthermore, this review explores potential future directions in the field, aiming to provide valuable insights to inform and enhance the clinical management of pancreatic cancer.
Collapse
Affiliation(s)
- Wenyu Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Jingzheng Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Wei Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- Department of Gastroenterology, Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- Department of Gastroenterology, Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| |
Collapse
|
21
|
Antony A, Mukherjee S, Bi Y, Collisson EA, Nagaraj M, Murlidhar M, Wallace MB, Goenka AH. AI-Driven insights in pancreatic cancer imaging: from pre-diagnostic detection to prognostication. Abdom Radiol (NY) 2024:10.1007/s00261-024-04775-x. [PMID: 39738571 DOI: 10.1007/s00261-024-04775-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related deaths in the United States, largely due to its poor five-year survival rate and frequent late-stage diagnosis. A significant barrier to early detection even in high-risk cohorts is that the pancreas often appears morphologically normal during the pre-diagnostic phase. Yet, the disease can progress rapidly from subclinical stages to widespread metastasis, undermining the effectiveness of screening. Recently, artificial intelligence (AI) applied to cross-sectional imaging has shown significant potential in identifying subtle, early-stage changes in pancreatic tissue that are often imperceptible to the human eye. Moreover, AI-driven imaging also aids in the discovery of prognostic and predictive biomarkers, essential for personalized treatment planning. This article uniquely integrates a critical discussion on AI's role in detecting visually occult PDAC on pre-diagnostic imaging, addresses challenges of model generalizability, and emphasizes solutions like standardized datasets and clinical workflows. By focusing on both technical advancements and practical implementation, this article provides a forward-thinking conceptual framework that bridges current gaps in AI-driven PDAC research.
Collapse
Affiliation(s)
- Ajith Antony
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Yan Bi
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Eric A Collisson
- Department of Medical Oncology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Madhu Nagaraj
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Michael B Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Ajit H Goenka
- Department of Radiology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
22
|
Wu J, Tang L, Zheng F, Chen X, Li L. A review of the last decade: pancreatic cancer and type 2 diabetes. Arch Physiol Biochem 2024; 130:660-668. [PMID: 37646618 DOI: 10.1080/13813455.2023.2252204] [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: 06/16/2023] [Revised: 08/04/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
Pancreatic cancer (PC) is a prevalent gastrointestinal tumour known for its high degree of malignancy, resulting in a mere 10% five-year survival rate for most patients. Over the past decade, a growing body of research has shed light on the intricate bidirectional association between PC and Type 2 diabetes (T2DM). The collection of PC- and T2DM-related articles is derived from two comprehensive databases, namely WOS (Web of Science Core Collection) and CNKI (China National Knowledge Infrastructure). This article discusses the last 10 years of research trends in PC and T2DM and explores their potential regulatory relationship as well as related medications.
Collapse
Affiliation(s)
- Jiaqi Wu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Nursing, Southern Medical University, Guangzhou, China
| | - Liang Tang
- Department of General Medicine, Zhuzhou Central Hospital, Zhuzhou, China
| | - Feng Zheng
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Xun Chen
- Department of the Trauma center, Zhuzhou Central Hospital, Zhuzhou, China
- Department of hepatobiliary surgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Lei Li
- Department of Pathology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
23
|
Meziani J, de Jong JG, Fuhler GM, Koopmann BD, Levink IJ, Fockens P, Vleggaar FP, Bruno MJ, Cahen DL. Assessment of Glucose and HbA1c Monitoring in a Pancreatic Cancer Surveillance Program for High-Risk Individuals. Clin Transl Gastroenterol 2024; 15:e00777. [PMID: 39413349 PMCID: PMC11671095 DOI: 10.14309/ctg.0000000000000777] [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: 04/11/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
INTRODUCTION Several studies suggest that new-onset diabetes mellitus is an early manifestation of pancreatic ductal adenocarcinoma (PDAC). Therefore, the International Cancer of the Pancreas Screening Consortium recommends glucose and hemoglobin A1c (HbA1c) monitoring in high-risk individuals (HRIs) undergoing surveillance. However, evidence that such monitoring improves PDAC detection is lacking. Our aim was to investigate the association between serum glucose and HbA1c values and the development of PDAC in HRIs undergoing surveillance. METHODS Participants were recruited from the familial pancreatic cancer surveillance cohort, which follows hereditary predisposed HRIs yearly by magnetic resonance imaging and/or endoscopic ultrasound and blood sampling. Those who underwent fasting glucose and/or HbA1c monitoring at least once were eligible candidates. RESULTS Four hundred four HRIs met the inclusion criteria. During a median follow-up of 41 months (range 14-120), 9 individuals developed PDAC and 4 (without PDAC) were diagnosed with new-onset diabetes mellitus. Glucose levels ranged from 3.4 to 10.7 mmol/L (mean 5.6 ± 0.7) and HbA1c levels from 25 to 68 mmol/mol (mean 37.7 ± 4.1). The mean values did not differ significantly between PDAC cases and controls. The percentage of individuals with at least one elevated value were comparable between PDAC cases and controls for glucose (33% and 27%, P = 0.707) and HbA1c (22% and 14%, P = 0.623). No consistent glucose or HbA1c trends over time suggested a correlation with PDAC development. DISCUSSION In this HRI surveillance cohort, measuring glucose and HbA1c values did not contribute to PDAC detection. Larger and longer-term studies are needed to determine the final role of glucose and HbA1c monitoring in PDAC surveillance.
Collapse
Affiliation(s)
- Jihane Meziani
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jedidja G.Y. de Jong
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Gwenny M. Fuhler
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Brechtje D.M. Koopmann
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Iris J.M. Levink
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Paul Fockens
- Department of Gastroenterology & Hepatology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Frank P. Vleggaar
- Department of Gastroenterology & Hepatology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marco J. Bruno
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Djuna L. Cahen
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
24
|
Smith LM, Mahoney DW, Bamlet WR, Yu F, Liu S, Goggins MG, Darabi S, Majumder S, Wang QL, Coté GA, Demeure MJ, Zhang Z, Srivastava S, Chawla A, Izmirlian G, Olson JE, Wolpin BM, Genkinger JM, Zaret KS, Brand R, Koay EJ, Oberg AL. Early detection of pancreatic cancer: Study design and analytical considerations in biomarker discovery and early phase validation studies. Pancreatology 2024; 24:1265-1279. [PMID: 39516175 PMCID: PMC11780679 DOI: 10.1016/j.pan.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/05/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease that is challenging to detect at an early stage. Biomarkers are needed that can detect PDAC early in the course of disease when interventions lead to the best outcomes. We highlight study design and statistical considerations that inform pancreatic cancer early detection biomarker evaluation. METHODS We describe experimental design strategies in this setting useful for streamlining biomarker evaluation at each Early Detection Research Network (EDRN) phase of biomarker development. We break the early EDRN phases into sub-phases, proposing objectives, study design strategies, and biomarker performance benchmarks. RESULTS The goal of early detection in populations at high-risk of PDAC is described. Evaluating biomarker behavior in patients under surveillance without disease can winnow candidate biomarkers. Potential resources for biomarker validation studies are described. CONCLUSIONS Multisite and multidisciplinary collaboration can facilitate study design strategies in this lethal but low incidence disease and streamline the path from biomarker discovery to clinical use. Improvements in analytical and experimental design methods could help accelerate biomarker evaluation through the phases of biomarker development.
Collapse
Affiliation(s)
- Lynette M Smith
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Douglas W Mahoney
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - William R Bamlet
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Fang Yu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Suyu Liu
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael G Goggins
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sourat Darabi
- Hoag Family Cancer Institute, Newport Beach, CA, USA
| | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Qiao-Li Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Gregory A Coté
- Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | | | - Zhen Zhang
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Akhil Chawla
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Janet E Olson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jeanine M Genkinger
- Department of Epidemiology, Mailman School of Public Health, Columbia University, NY, NY, USA
| | - Kenneth S Zaret
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Randall Brand
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eugene J Koay
- Department of Gastrointestinal Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ann L Oberg
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
25
|
Lee GH, Kim YH, Woo SM, Lee WJ, Han SS, Park SJ, Price S, Tembo P, Hébert JR, Kim MK. The Impact of the Dietary Inflammatory Index, Fasting Blood Glucose, and Smoking Status on the Incidence and Survival of Pancreatic Cancer: A Retrospective Case-Control Study and a Prospective Study. Nutrients 2024; 16:3941. [PMID: 39599726 PMCID: PMC11597200 DOI: 10.3390/nu16223941] [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: 10/10/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Pancreatic cancer (PC), a highly malignant cancer with a poor diagnosis, may be influenced by diet-related inflammation. This study examined the association between dietary inflammatory index (DII) scores and the incidence and prognosis of PC in Korea. METHODS A total of 55 patients with PC were matched with 280 healthy controls (HCs) by age and sex. We also analyzed the combined effects of DII scores and fasting blood glucose (FBG) levels or smoking status on the risk of PC and performed a survival analysis using the Cox proportional hazards method. RESULTS The DII scores were higher in the patients with PC than those in HCs (odds ratio [OR] = 3.36, confidence interval [CI] = 1.16-9.73, p = 0.03), and the effect was larger in women (OR = 6.13, CI = 1.11-33.82, p = 0.04). A high DII score was jointly associated with FBG ≥ 126 mg/dL in raising PC risk [OR = 32.5, relative excess risk due to interaction/synergy (RERI/S) index = 24.2/4.34, p-interaction = 0.04], indicating a multiplicative interaction. A high DII score combined with ex/current smoker status increased PC risk through an additive interaction (RERI/S = 1.01/1.54, p-interaction = 0.76). However, DII scores did not influence disease-free survival. CONCLUSIONS The consumption of an anti-inflammatory diet, coupled with maintaining normal FBG levels and abstaining from smoking, may help reduce the risk of PC by mitigating pancreatic inflammation.
Collapse
Affiliation(s)
- Ga Hyun Lee
- Cancer Epidemiology Branch, Division of Cancer Epidemiology and Prevention, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Republic of Korea; (G.H.L.); (Y.H.K.)
| | - Yeon Hee Kim
- Cancer Epidemiology Branch, Division of Cancer Epidemiology and Prevention, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Republic of Korea; (G.H.L.); (Y.H.K.)
| | - Sang Myung Woo
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Republic of Korea; (S.M.W.); (W.J.L.); (S.-S.H.); (S.-J.P.)
| | - Woo Jin Lee
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Republic of Korea; (S.M.W.); (W.J.L.); (S.-S.H.); (S.-J.P.)
| | - Sung-Sik Han
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Republic of Korea; (S.M.W.); (W.J.L.); (S.-S.H.); (S.-J.P.)
| | - Sang-Jae Park
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Republic of Korea; (S.M.W.); (W.J.L.); (S.-S.H.); (S.-J.P.)
| | - Sherry Price
- Department of Epidemiology and Biostatistics and Cancer Prevention and Control Program, University of South Carolina, Columbia, SC 29208, USA; (S.P.); (P.T.); (J.R.H.)
| | - Penias Tembo
- Department of Epidemiology and Biostatistics and Cancer Prevention and Control Program, University of South Carolina, Columbia, SC 29208, USA; (S.P.); (P.T.); (J.R.H.)
| | - James R. Hébert
- Department of Epidemiology and Biostatistics and Cancer Prevention and Control Program, University of South Carolina, Columbia, SC 29208, USA; (S.P.); (P.T.); (J.R.H.)
- Department of Nutrition, Connecting Health Innovations LLC, Columbia, SC 29201, USA
| | - Mi Kyung Kim
- Cancer Epidemiology Branch, Division of Cancer Epidemiology and Prevention, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Republic of Korea; (G.H.L.); (Y.H.K.)
| |
Collapse
|
26
|
Wlodarczyk B, Durko L, Walczak K, Talar-Wojnarowska R, Malecka-Wojciesko E. Select Endocrine Disorders and Exosomes in Early PDAC Diagnosis. Int J Mol Sci 2024; 25:12159. [PMID: 39596226 PMCID: PMC11594802 DOI: 10.3390/ijms252212159] [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: 09/16/2024] [Revised: 11/01/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
Disturbances in carbohydrate metabolism are suggested to be the early symptoms of pancreatic ductal adenocarcinoma (PDAC). The accumulated data suggests that endocrine function-related biomarkers may represent a breakthrough in the early detection of PDAC. Factors which may predispose one to the development of PDAC are insulin resistance and hyperinsulinemia. Elevated insulin levels induce the onset of carcinogenesis by altering the differentiation and function of islet cells through stimulating growth factors, including insulin-like growth factors (IGFs). Impaired β cell function, along with the impact of PDAC-released factors (e.g., adrenomedullin (ADM), IGF-1, and macrophage inhibitory factor (MIF) on pancreatic islets, may contribute to the induction of diabetes associated with PDAC. Recently, exosomes have attracted worldwide attention due to their role in varied features of cell function, particularly in cancer progression. Exosomes comprise of small extracellular vesicles produced by almost all cells. These vesicles contain a vast array of biomolecules, including proteins and microRNAs. Exosomes participate in cancer growth and promote angiogenesis. They promote tumorigenesis and metastasis, and are associated with the acquisition of cancer cells resistant to chemotherapy. Data have been accumulating recently on the role of exosomes in the rapid recognition, prognosis and potential therapy of pancreatic cancer.
Collapse
Affiliation(s)
- Barbara Wlodarczyk
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-153 Lodz, Poland
| | - Lukasz Durko
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-153 Lodz, Poland
| | - Konrad Walczak
- Department of Internal Diseases and Nephrodiabetology, Medical University of Lodz, 90-549 Lodz, Poland
| | | | - Ewa Malecka-Wojciesko
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-153 Lodz, Poland
| |
Collapse
|
27
|
Gong J, Li X, Feng Z, Lou J, Pu K, Sun Y, Hu S, Zhou Y, Song T, Shangguan M, Zhang K, Lu W, Dong X, Wu J, Zhu H, He Q, Xu H, Wu Y. Sorcin can trigger pancreatic cancer-associated new-onset diabetes through the secretion of inflammatory cytokines such as serpin E1 and CCL5. Exp Mol Med 2024; 56:2535-2547. [PMID: 39516378 PMCID: PMC11612510 DOI: 10.1038/s12276-024-01346-4] [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: 11/18/2023] [Revised: 07/28/2024] [Accepted: 08/19/2024] [Indexed: 11/16/2024] Open
Abstract
A rise in blood glucose is an early warning sign of underlying pancreatic cancer (PC) and may be an indicator of genetic events in PC progression. However, there is still a lack of mechanistic research on pancreatic cancer-associated new-onset diabetes (PCAND). In the present study, we identified a gene SRI, which possesses a SNP with the potential to distinguish PCAND and Type 2 diabetes mellitus (T2DM), by machine learning on the basis of the UK Biobank database. In vitro and in vivo, sorcin overexpression induced pancreatic β-cell dysfunction. Sorcin can form a positive feedback loop with STAT3 to increase the transcription of serpin E1 and CCL5, which may directly induce β-cell dysfunction. In 88 biopsies, the expression of sorcin was elevated in PC tissues, especially in PCAND samples. Furthermore, plasma serpin E1 levels are higher in peripheral blood samples from PCAND patients than in those from T2DM patients. In conclusion, sorcin may be the key driver in PCAND, and further study on the sorcin-STAT3-serpin E1/CCL5 signaling axis may help us better understand the pathogenesis of PCAND and identify potential biomarkers.
Collapse
Affiliation(s)
- Jiali Gong
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Xiawei Li
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Zengyu Feng
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianyao Lou
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kaiyue Pu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yongji Sun
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Sien Hu
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Yizhao Zhou
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tianyu Song
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Meihua Shangguan
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kai Zhang
- School of Public Health and Eye Center The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Wenjie Lu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xin Dong
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jian Wu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Wenzhou, Zhejiang University, Wenzhou, Zhejiang, China
| | - Hong Zhu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiaojun He
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
- Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, Zhejiang, China.
| | - Hongxia Xu
- Innovation Institute for Artificial Intelligence in Medicine and Liangzhu Laboratory, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Yulian Wu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China.
| |
Collapse
|
28
|
Haab B, Qian L, Staal B, Jain M, Fahrmann J, Worthington C, Prosser D, Velokokhatnaya L, Lopez C, Tang R, Hurd MW, Natarajan G, Kumar S, Smith L, Hanash S, Batra SK, Maitra A, Lokshin A, Huang Y, Brand RE. A rigorous multi-laboratory study of known PDAC biomarkers identifies increased sensitivity and specificity over CA19-9 alone. Cancer Lett 2024; 604:217245. [PMID: 39276915 PMCID: PMC11808537 DOI: 10.1016/j.canlet.2024.217245] [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: 05/10/2024] [Revised: 08/24/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
A blood test that enables surveillance for early-stage pancreatic ductal adenocarcinoma (PDAC) is an urgent need. Independent laboratories have reported PDAC biomarkers that could improve biomarker performance over CA19-9 alone, but the performance of the previously reported biomarkers in combination is not known. Therefore, we conducted a coordinated case/control study across multiple laboratories using common sets of blinded training and validation samples (132 and 295 plasma samples, respectively) from PDAC patients and non-PDAC control subjects representing conditions under which surveillance occurs. We analyzed the training set to identify candidate biomarker combination panels using biomarkers across laboratories, and we applied the fixed panels to the validation set. The panels identified in the training set, CA19-9 with CA199.STRA, LRG1, TIMP-1, TGM2, THSP2, ANG, and MUC16.STRA, achieved consistent performance in the validation set. The panel of CA19-9 with the glycan biomarker CA199.STRA improved sensitivity from 0.44 with 0.98 specificity for CA19-9 alone to 0.71 with 0.98 specificity (p < 0.001, 1000-fold bootstrap). Similarly, CA19-9 combined with the protein biomarker LRG1 and CA199.STRA improved specificity from 0.16 with 0.94 sensitivity for CA19-9 to 0.65 with 0.89 sensitivity (p < 0.001, 1000-fold bootstrap). We further validated significantly improved performance using biomarker panels that did not include CA19-9. This study establishes the effectiveness of a coordinated study of previously discovered biomarkers and identified panels of those biomarkers that significantly increased the sensitivity and specificity of early-stage PDAC detection in a rigorous validation trial.
Collapse
Affiliation(s)
- Brian Haab
- Van Andel Institute, 333 Bostwick NE, Grand Rapids, MI, 49503, USA.
| | - Lu Qian
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 19024, USA
| | - Ben Staal
- Van Andel Institute, 333 Bostwick NE, Grand Rapids, MI, 49503, USA
| | - Maneesh Jain
- University of Nebraska Medical Center, 42nd and Emile Streets, Omaha, NE, 68198, USA
| | - Johannes Fahrmann
- MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Christine Worthington
- University of Pittsburgh Medical Center, 200 Lothrop St., Pittsburgh, PA, 15213-2582, USA
| | - Denise Prosser
- University of Pittsburgh Medical Center, 200 Lothrop St., Pittsburgh, PA, 15213-2582, USA
| | | | - Camden Lopez
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 19024, USA
| | - Runlong Tang
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 19024, USA
| | - Mark W Hurd
- MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | | | - Sushil Kumar
- University of Nebraska Medical Center, 42nd and Emile Streets, Omaha, NE, 68198, USA
| | - Lynette Smith
- University of Nebraska Medical Center, 42nd and Emile Streets, Omaha, NE, 68198, USA
| | - Sam Hanash
- MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Surinder K Batra
- University of Nebraska Medical Center, 42nd and Emile Streets, Omaha, NE, 68198, USA
| | - Anirban Maitra
- MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Anna Lokshin
- University of Pittsburgh Medical Center, 200 Lothrop St., Pittsburgh, PA, 15213-2582, USA
| | - Ying Huang
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA, 19024, USA
| | - Randall E Brand
- University of Pittsburgh Medical Center, 200 Lothrop St., Pittsburgh, PA, 15213-2582, USA.
| |
Collapse
|
29
|
Sun Y, Hu C, Hu S, Xu H, Gong J, Wu Y, Fan Y, Lv C, Song T, Lou J, Zhang K, Wu J, Li X, Wu Y. Predicting Pancreatic Cancer in New-Onset Diabetes Cohort Using a Novel Model With Integrated Clinical and Genetic Indicators: A Large-Scale Prospective Cohort Study. Cancer Med 2024; 13:e70388. [PMID: 39526476 PMCID: PMC11551786 DOI: 10.1002/cam4.70388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/30/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
INTRODUCTION Individuals who develop new-onset diabetes have been identified as a high-risk cohort for pancreatic cancer (PC), exhibiting an incidence rate nearly 8 times higher than the general population. Hence, the targeted screening of this specific cohort presents a promising opportunity for early pancreatic cancer detection. We aimed to develop and validate a novel model capable of identifying high-risk individuals among those with new-onset diabetes. METHODS Employing the UK Biobank cohort, we focused on those developing new-onset diabetes during follow-up. Genetic and clinical characteristics available at registration were considered as candidate predictors. We conducted univariate regression analysis to identify potential indicators and used a 5-fold cross-validation method to select optimal predictors for model development. Five machine learning algorithms were used for model development. RESULTS Among 12,735 patients with new-onset diabetes, 100 (0.8%) were diagnosed with PC within 2 years. The final model (area under the curve, 0.897; 95% confidence interval, 0.865-0.929) included 5 clinical predictors and 24 single nucleotide polymorphisms. Two threshold cut-offs were established: 1.28% and 5.26%. The recommended 1.28% cut-off, based on model performance, reduces definitive testing to 13% of the total population while capturing 76% of PC cases. The high-risk threshold is 5.26%. Utilizing this threshold, only 2% of the population needs definitive testing, capturing nearly half of PC cases. CONCLUSIONS We, for the first time, combined clinical and genetic data to develop and validate a model to determine the risk of pancreatic cancer in patients with new-onset diabetes using machine learning algorithms. By reducing the number of unnecessary tests while ensuring that a substantial proportion of high-risk patients are identified, this tool has the potential to improve patient outcomes and optimize healthcare sources.
Collapse
Affiliation(s)
- Yongji Sun
- Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
| | - Chaowen Hu
- Polytechnic InstituteZhejiang UniversityHangzhouZhejiangChina
| | - Sien Hu
- Department of General SurgeryHangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical UniversityZhejiangHangzhouChina
| | - Hongxia Xu
- Innovation Institute for Artificial Intelligence in MedicineZhejiang UniversityHangzhouChina
| | - Jiali Gong
- Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer InstituteSecond Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiangChina
- Cancer CenterZhejiang UniversityHangzhouZhejiangChina
| | - Yixuan Wu
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Yiqun Fan
- Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer InstituteSecond Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiangChina
- Cancer CenterZhejiang UniversityHangzhouZhejiangChina
| | - Changming Lv
- Department of Surgery, Fourth Affiliated Hospital, International Institutes of MedicineZhejiang University School of MedicineZhejiangChina
- Institute of WenzhouZhejiang UniversityZhejiangChina
| | - Tianyu Song
- Department of Surgery, Fourth Affiliated Hospital, International Institutes of MedicineZhejiang University School of MedicineZhejiangChina
- Institute of WenzhouZhejiang UniversityZhejiangChina
| | - Jianyao Lou
- Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer InstituteSecond Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiangChina
- Cancer CenterZhejiang UniversityHangzhouZhejiangChina
| | - Kai Zhang
- School of Public Health and Eye CenterThe Second Affiliated Hospital, Zhejiang UniversityHangzhouChina
| | - Jian Wu
- Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Innovation Institute for Artificial Intelligence in MedicineZhejiang UniversityHangzhouChina
- Cancer CenterZhejiang UniversityHangzhouZhejiangChina
- Department of Surgery, Fourth Affiliated Hospital, International Institutes of MedicineZhejiang University School of MedicineZhejiangChina
- Institute of WenzhouZhejiang UniversityZhejiangChina
| | - Xiawei Li
- Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer InstituteSecond Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiangChina
- Cancer CenterZhejiang UniversityHangzhouZhejiangChina
- School of Public HealthZhejiang University School of MedicineZhejiangHangzhouChina
| | - Yulian Wu
- Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer InstituteSecond Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiangChina
- Cancer CenterZhejiang UniversityHangzhouZhejiangChina
- Department of Surgery, Fourth Affiliated Hospital, International Institutes of MedicineZhejiang University School of MedicineZhejiangChina
- Institute of WenzhouZhejiang UniversityZhejiangChina
| |
Collapse
|
30
|
Chen W, Zhou B, Luong TQ, Lustigova E, Xie F, Matrisian LM, Wu BU. Prediction of pancreatic cancer in patients with new onset hyperglycemia: A modified ENDPAC model. Pancreatology 2024; 24:1115-1122. [PMID: 39353843 DOI: 10.1016/j.pan.2024.09.015] [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: 06/19/2024] [Revised: 08/27/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND/OBJECTIVES The Enriching New-Onset Diabetes for Pancreatic Cancer (ENDPAC) model relies primarily on fasting glucose values. Health systems have increasingly shifted practice towards use of glycated hemoglobin (HbA1c) measurement. We modified the ENDPAC model using patients with new onset hyperglycemia. METHODS Four cohorts of patients 50-84 years of age with HbA1c results ≥6.2-6.5 % in 2011-2018 were identified. A combine cohort was formed. A widened eligibility criterion was applied to form additional four individual cohorts and one combined cohort. The primary outcome was the diagnosis of pancreatic cancer within 3 years after the first elevated HbA1c testing. The performance of the modified ENDPAC model was evaluated by AUC, sensitivity, positive predictive value, cases detected, and total number of patients screened. RESULTS The individual and combined cohorts consisted of 39,001-79,060 and 69,334-92,818 patients, respectively (mean age 63.5-65.0 years). The three-year PC incidence rates were 0.47%-0.54 %. The AUC measures were in the range of 0.75-0.77 for the individual cohorts and 0.75 for the combined cohorts. When the four individual cohorts were combined, more PC cases can be identified (149 by the combined vs. 113-116 by individual cohorts when risk score was 5+). Performance measures were compromised in nonwhites. Asian and Pacific islanders had lower sensitivity compared to other racial and ethnic groups (29 % vs. 50-60 %) when risk score was 5+. CONCLUSIONS The modified ENDPAC model targets a broader population and thus identifies more high-risk patients for cancer screening. The differential performance needs to be considered when the model is applied to non-white population.
Collapse
Affiliation(s)
- Wansu Chen
- Kaiser Permanente Southern California Research and Evaluation, 100 S. Los Robles Ave, Pasadena, CA, USA.
| | - Botao Zhou
- Kaiser Permanente Southern California Research and Evaluation, 100 S. Los Robles Ave, Pasadena, CA, USA
| | - Tiffany Q Luong
- Kaiser Permanente Southern California Research and Evaluation, 100 S. Los Robles Ave, Pasadena, CA, USA
| | - Eva Lustigova
- Kaiser Permanente Southern California Research and Evaluation, 100 S. Los Robles Ave, Pasadena, CA, USA
| | - Fagen Xie
- Kaiser Permanente Southern California Research and Evaluation, 100 S. Los Robles Ave, Pasadena, CA, USA
| | - Lynn M Matrisian
- Pancreatic Cancer Action Network, 1500 Rosecrans Ave, Suite 200, Manhattan Beach, CA, USA
| | - Bechien U Wu
- Center for Pancreatic Care, Department of Gastroenterology, Los Angeles Medical Center, Southern California Permanente Medical Group, 4867 West Sunset Blvd, Los Angeles, CA, USA
| |
Collapse
|
31
|
García García de Paredes A, Martínez Moneo E, Lariño-Noia J, Earl J. Pancreatic cancer screening in high-risk individuals. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2024; 116:519-522. [PMID: 39087662 DOI: 10.17235/reed.2024.10635/2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
The incidence of pancreatic cancer is increasing, although globally it represents less than 3% of all cancers. Despite advances in medical and surgical management, survival rates have not significantly improved in recent years. Consequently, pancreatic cancer, though relatively uncommon, is the third leading cause of cancer-related deaths. This is primarily due to the disease´s late detection. Symptoms appear late and are nonspecific, and over 80% of cases are diagnosed at an advanced stage and unsuitable for curative surgery, resulting in a five-year survival rate below 10%. However, the exceptional cases that are diagnosed early show five-year survival rates exceeding 80%. Therefore, one of the keys to improving pancreatic cancer prognosis lies in early detection, making screening in high-risk individuals a potentially crucial strategy.
Collapse
Affiliation(s)
| | | | | | - Julie Earl
- Biomarkers and Personalized Approach to Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Munnings R, Gibbs P, Lee B. Evolution of Liquid Biopsies for Detecting Pancreatic Cancer. Cancers (Basel) 2024; 16:3335. [PMID: 39409954 PMCID: PMC11475855 DOI: 10.3390/cancers16193335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy characterised by late diagnosis and poor prognosis. Despite advancements, current diagnostic and prognostic strategies remain limited. Liquid biopsy techniques, including circulating tumour DNA (ctDNA), circulating tumour cells (CTCs), circulating tumour exosomes, and proteomics, offer potential solutions to improve PDAC diagnosis, prognostication, and management. A systematic search of Ovid MEDLINE identified studies published between 2019 and 2024, focusing on liquid biopsy biomarkers for PDAC. A total of 49 articles were included. ctDNA research shows some promise in diagnosing and prognosticating PDAC, especially through detecting mutant KRAS in minimal residual disease assays. CTC analyses had low sensitivity for early-stage PDAC and inconsistent prognostic results across subpopulations. Exosomal studies revealed diverse biomarkers with some diagnostic and prognostic potential. Proteomics, although relatively novel, has demonstrated superior accuracy in PDAC diagnosis, including early detection, and notable prognostic capacity. Proteomics combined with CA19-9 analysis has shown the most promising results to date. An update on multi-cancer early detection testing, given its significance for population screening, is also briefly discussed. Liquid biopsy techniques offer promising avenues for improving PDAC diagnosis, prognostication, and management. In particular, proteomics shows considerable potential, yet further research is needed to validate existing findings and comprehensively explore the proteome using an unbiased approach.
Collapse
Affiliation(s)
- Ryan Munnings
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
- Department of Medical Education, Melbourne Medical School, Parkville, VIC 3052, Australia
| | - Peter Gibbs
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
- Western Health, Footscray, VIC 3011, Australia
| | - Belinda Lee
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
- Peter MacCallum Cancer Centre, Parkville, VIC 3052, Australia
- Northern Health, Epping, VIC 3076, Australia
| |
Collapse
|
35
|
Klatte DCF, Weston A, Ma Y, Sledge H, Bali A, Bolan C, Engels M, van Hooft JE, van Leerdam ME, Ouni A, Wallace MB, Bi Y. Temporal Trends in Body Composition and Metabolic Markers Prior to Diagnosis of Pancreatic Ductal Adenocarcinoma. Clin Gastroenterol Hepatol 2024; 22:1830-1838.e9. [PMID: 38703880 DOI: 10.1016/j.cgh.2024.03.038] [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: 12/20/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND & AIMS Changes in body composition and metabolic factors may serve as biomarkers for the early detection of pancreatic ductal adenocarcinoma (PDAC). The aim of this study was to capture the longitudinal changes in body composition and metabolic factors before diagnosis of PDAC. METHODS We performed a retrospective cohort study in which all patients (≥18 years) diagnosed with PDAC from 2002 to 2021 were identified. We collected all abdominal computed tomography scans and 10 different blood-based biomarkers up to 36 months before diagnosis. We applied a fully automated abdominal segmentation algorithm previously developed by our group for 3-dimensional quantification of body composition on computed tomography scans. Longitudinal trends of body composition and blood-based biomarkers before PDAC diagnosis were estimated using linear mixed models, compared across different time windows, and visualized using spline regression. RESULTS We included 1690 patients in body composition analysis, of whom 516 (30.5%) had ≥2 prediagnostic computed tomography scans. For analysis of longitudinal trends of blood-based biomarkers, 3332 individuals were included. As an early manifestation of PDAC, we observed a significant decrease in visceral and subcutaneous adipose tissue (β = -1.94 [95% confidence interval (CI), -2.39 to -1.48] and β = -2.59 [95% CI, -3.17 to -2.02]) in area (cm2)/height (m2) per 6 months closer to diagnosis, accompanied by a decrease in serum lipids (eg, low-density lipoprotein [β = -2.83; 95% CI, -3.31 to -2.34], total cholesterol [β = -2.69; 95% CI, -3.18 to -2.20], and triglycerides [β = -1.86; 95% CI, -2.61 to -1.11]), and an increase in blood glucose levels. Loss of muscle tissue and bone volume was predominantly observed in the last 6 months before diagnosis. CONCLUSIONS This study identified significant alterations in a variety of soft tissue and metabolic markers that occur in the development of PDAC. Early recognition of these metabolic changes may provide an opportunity for early detection.
Collapse
Affiliation(s)
- Derk C F Klatte
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Alexander Weston
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Yaohua Ma
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Hanna Sledge
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Aman Bali
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | - Candice Bolan
- Department of Radiology, Mayo Clinic, Jacksonville, Florida
| | - Megan Engels
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeanin E van Hooft
- 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; Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ahmed Ouni
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | - Michael B Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | - Yan Bi
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
36
|
Patterson L, Toledo FGS, Maitra A, Chari ST. Pancreatic Cancer-Induced Metabolic Dysregulation Syndrome: Clinical Profile, Proposed Mechanisms, and Unanswered Questions. Gastroenterology 2024:S0016-5085(24)05412-X. [PMID: 39222716 DOI: 10.1053/j.gastro.2024.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 08/02/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Affiliation(s)
- LaNisha Patterson
- Department of Pathology and Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frederico G S Toledo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anirban Maitra
- Department of Pathology and Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Suresh T Chari
- Department of Gastroenterology, Hepatology, and Nutrition, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
37
|
Grigorescu RR, Husar-Sburlan IA, Gheorghe C. Pancreatic Cancer: A Review of Risk Factors. Life (Basel) 2024; 14:980. [PMID: 39202722 PMCID: PMC11355429 DOI: 10.3390/life14080980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Pancreatic adenocarcinoma is one of the most lethal types of gastrointestinal cancer despite the latest medical advances. Its incidence has continuously increased in recent years in developed countries. The location of the pancreas can result in the initial symptoms of neoplasia being overlooked, which can lead to a delayed diagnosis and a subsequent reduction in the spectrum of available therapeutic options. The role of modifiable risk factors in pancreatic cancer has been extensively studied in recent years, with smoking and alcohol consumption identified as key contributors. However, the few screening programs that have been developed focus exclusively on genetic factors, without considering the potential impact of modifiable factors on disease occurrence. Thus, fully understanding and detecting the risk factors for pancreatic cancer represents an important step in the prevention and early diagnosis of this type of neoplasia. This review reports the available evidence on different risk factors and identifies the areas that could benefit the most from additional studies.
Collapse
Affiliation(s)
- Raluca Roxana Grigorescu
- Gastroenterology Department, “Sfanta Maria” Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | | | - Cristian Gheorghe
- Center for Digestive Disease and Liver Transplantation, Fundeni Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
38
|
Kaul M, Sanin AY, Shi W, Janiak C, Kahlert UD. Nanoformulation of dasatinib cannot overcome therapy resistance of pancreatic cancer cells with low LYN kinase expression. Pharmacol Rep 2024; 76:793-806. [PMID: 38739359 PMCID: PMC11294441 DOI: 10.1007/s43440-024-00600-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: 11/02/2023] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/14/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most difficult to treat tumors. The Src (sarcoma) inhibitor dasatinib (DASA) has shown promising efficacy in preclinical studies of PDAC. However, clinical confirmation could not be achieved. Overall, our aim was to deliver arguments for the possible reinitiating clinical testing of this compound in a biomarker-stratifying therapy trial for PDAC patients. We tested if the nanofunctionalization of DASA can increase the drug efficacy and whether certain Src members can function as clinical predictive biomarkers. METHODS Methods include manufacturing of poly(vinyl alcohol) stabilized gold nanoparticles and their drug loading, dynamic light scattering, transmission electron microscopy, thermogravimetric analysis, Zeta potential measurement, sterile human cell culture, cell growth quantification, accessing and evaluating transcriptome and clinical data from molecular tumor dataset TCGA, as well as various statistical analyses. RESULTS We generated homo-dispersed nanofunctionalized DASA as an AuNP@PVA-DASA conjugate. The composite did not enhance the anti-growth effect of DASA on PDAC cell lines. The cell model with high LYN expression showed the strongest response to the therapy. We confirm deregulated Src kinetome activity as a prevalent feature of PDAC by revealing mRNA levels associated with higher malignancy grade of tumors. BLK (B lymphocyte kinase) expression predicts shorter overall survival of diabetic PDAC patients. CONCLUSIONS Nanofunctionalization of DASA needs further improvement to overcome the therapy resistance of PDAC. LYN mRNA is augmented in tumors with higher malignancy and can serve as a predictive biomarker for the therapy resistance of PDAC cells against DASA. Studying the biological roles of BLK might help to identify underlying molecular mechanisms associated with PDAC in diabetic patients.
Collapse
Affiliation(s)
- Marilyn Kaul
- Institute for Inorganic and Structural Chemistry, Heinrich-Heine-University Düsseldorf, 40204, Düsseldorf, Germany
| | - Ahmed Y Sanin
- Molecular and Experimental Surgery, University Clinic for General-, Visceral-, Vascular- and Transplant Surgery, Faculty of Medicine, Otto-Von-Guericke-University Magdeburg, 39120, Magdeburg, Germany
| | - Wenjie Shi
- Molecular and Experimental Surgery, University Clinic for General-, Visceral-, Vascular- and Transplant Surgery, Faculty of Medicine, Otto-Von-Guericke-University Magdeburg, 39120, Magdeburg, Germany
| | - Christoph Janiak
- Institute for Inorganic and Structural Chemistry, Heinrich-Heine-University Düsseldorf, 40204, Düsseldorf, Germany.
| | - Ulf D Kahlert
- Molecular and Experimental Surgery, University Clinic for General-, Visceral-, Vascular- and Transplant Surgery, Faculty of Medicine, Otto-Von-Guericke-University Magdeburg, 39120, Magdeburg, Germany.
- Institute for Quality Assurance in Operative Medicine, Otto-Von-Guericke University at Magdeburg, Magdeburg, Germany.
| |
Collapse
|
39
|
Mishra AK, Chong B, Arunachalam SP, Oberg AL, Majumder S. Machine Learning Models for Pancreatic Cancer Risk Prediction Using Electronic Health Record Data-A Systematic Review and Assessment. Am J Gastroenterol 2024; 119:1466-1482. [PMID: 38752654 PMCID: PMC11296923 DOI: 10.14309/ajg.0000000000002870] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/06/2024] [Indexed: 06/20/2024]
Abstract
INTRODUCTION Accurate risk prediction can facilitate screening and early detection of pancreatic cancer (PC). We conducted a systematic review to critically evaluate effectiveness of machine learning (ML) and artificial intelligence (AI) techniques applied to electronic health records (EHR) for PC risk prediction. METHODS Ovid MEDLINE(R), Ovid EMBASE, Ovid Cochrane Central Register of Controlled Trials, Ovid Cochrane Database of Systematic Reviews, Scopus, and Web of Science were searched for articles that utilized ML/AI techniques to predict PC, published between January 1, 2012, and February 1, 2024. Study selection and data extraction were conducted by 2 independent reviewers. Critical appraisal and data extraction were performed using the CHecklist for critical Appraisal and data extraction for systematic Reviews of prediction Modelling Studies checklist. Risk of bias and applicability were examined using prediction model risk of bias assessment tool. RESULTS Thirty studies including 169,149 PC cases were identified. Logistic regression was the most frequent modeling method. Twenty studies utilized a curated set of known PC risk predictors or those identified by clinical experts. ML model discrimination performance (C-index) ranged from 0.57 to 1.0. Missing data were underreported, and most studies did not implement explainable-AI techniques or report exclusion time intervals. DISCUSSION AI/ML models for PC risk prediction using known risk factors perform reasonably well and may have near-term applications in identifying cohorts for targeted PC screening if validated in real-world data sets. The combined use of structured and unstructured EHR data using emerging AI models while incorporating explainable-AI techniques has the potential to identify novel PC risk factors, and this approach merits further study.
Collapse
Affiliation(s)
- Anup Kumar Mishra
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Bradford Chong
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | - Ann L. Oberg
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Shounak Majumder
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
40
|
Rajagopalan A, Aroori S, Russell TB, Labib PL, Ausania F, Pando E, Roberts KJ, Kausar A, Mavroeidis VK, Marangoni G, Thomasset SC, Frampton AE, Lykoudis P, Maglione M, Alhaboob N, Bari H, Smith AM, Spalding D, Srinivasan P, Davidson BR, Bhogal RH, Dominguez I, Thakkar R, Gomez D, Silva MA, Lapolla P, Mingoli A, Porcu A, Shah NS, Hamady ZZR, Al-Sarrieh B, Serrablo A, Croagh D. Five-year recurrence/survival after pancreatoduodenectomy for pancreatic adenocarcinoma: does pre-existing diabetes matter? Results from the Recurrence After Whipple's (RAW) study. HPB (Oxford) 2024; 26:981-989. [PMID: 38755085 DOI: 10.1016/j.hpb.2024.04.010] [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: 09/09/2023] [Revised: 03/27/2024] [Accepted: 04/19/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Diabetes mellitus (DM) has a complex relationship with pancreatic cancer. This study examines the impact of preoperative DM, both recent-onset and pre-existing, on long-term outcomes following pancreatoduodenectomy (PD) for pancreatic ductal adenocarcinoma (PDAC). METHODS Data were extracted from the Recurrence After Whipple's (RAW) study, a multi-centre cohort of PD for pancreatic head malignancy (2012-2015). Recurrence and five-year survival rates of patients with DM were compared to those without, and subgroup analysis performed to compare patients with recent-onset DM (less than one year) to patients with established DM. RESULTS Out of 758 patients included, 187 (24.7%) had DM, of whom, 47 of the 187 (25.1%) had recent-onset DM. There was no difference in the rate of postoperative pancreatic fistula (DM: 5.9% vs no DM 9.8%; p = 0.11), five-year survival (DM: 24.1% vs no DM: 22.9%; p = 0.77) or five-year recurrence (DM: 71.7% vs no DM: 67.4%; p = 0.32). There was also no difference between patients with recent-onset DM and patients with established DM in postoperative outcomes, recurrence, or survival. CONCLUSION We found no difference in five-year recurrence and survival between diabetic patients and those without diabetes. Patients with pre-existing DM should be evaluated for PD on a comparable basis to non-diabetic patients.
Collapse
Affiliation(s)
| | | | | | - Peter L Labib
- University Hospitals Plymouth NHS Trust, Plymouth, UK
| | | | | | - Keith J Roberts
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | | | | | | | | | | | | | | | - Hassaan Bari
- Shaukat Khanum Memorial Cancer Hospital, Lahore, Pakistan
| | | | | | | | | | | | - Ismael Dominguez
- Salvador Zubiran National Institute of Health Sciences and Nutrition, Mexico City, Mexico
| | - Rohan Thakkar
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Dhanny Gomez
- Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Michael A Silva
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Andrea Mingoli
- Policlinico Umberto I University Hospital Sapienza, Rome, Italy
| | - Alberto Porcu
- Azienda Ospedaliero Universitaria di Sassari, Sassari, Italy
| | - Nehal S Shah
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Zaed Z R Hamady
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | | | | |
Collapse
|
41
|
Wang L, Levinson R, Mezzacappa C, Katona BW. Review of the cost-effectiveness of surveillance for hereditary pancreatic cancer. Fam Cancer 2024; 23:351-360. [PMID: 38795221 PMCID: PMC11255025 DOI: 10.1007/s10689-024-00392-1] [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: 12/01/2023] [Accepted: 04/16/2024] [Indexed: 05/27/2024]
Abstract
Individuals with hereditary pancreatic cancer risk include high risk individuals (HRIs) with germline genetic susceptibility to pancreatic cancer (PC) and/or a strong family history of PC. Previously, studies have shown that PC surveillance in HRIs can downstage PC diagnosis and extend survival leading to pancreatic surveillance being recommended for certain HRIs. However, the optimal surveillance strategy remains uncertain, including which modalities should be used for surveillance, how frequently should surveillance be performed, and which sub-groups of HRIs should undergo surveillance. Additionally, in the ideal world PC surveillance should also be cost-effective. Cost-effectiveness analysis is a valuable tool that can consider the costs, potential health benefits, and risks among various PC surveillance strategies. In this review, we summarize the cost-effectiveness of various PC surveillance strategies for HRIs for hereditary pancreatic cancer and provide potential avenues for future work in this field. Additionally, we include cost-effectiveness studies among individuals with new-onset diabetes (NoD), a high-risk group for sporadic PC, as a comparison.
Collapse
Affiliation(s)
- Louise Wang
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd. 751 South Pavilion, Philadelphia, PA, 19104, USA
| | - Rachel Levinson
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT, 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
|
42
|
Khan S, Bhushan B. Machine Learning Predicts Patients With New-onset Diabetes at Risk of Pancreatic Cancer. J Clin Gastroenterol 2024; 58:681-691. [PMID: 37522752 DOI: 10.1097/mcg.0000000000001897] [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: 04/04/2023] [Accepted: 06/22/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND New-onset diabetes represent a high-risk cohort to screen for pancreatic cancer. GOALS Develop a machine model to predict pancreatic cancer among patients with new-onset diabetes. STUDY A retrospective cohort of patients with new-onset diabetes was assembled from multiple health care networks in the United States. An XGBoost machine learning model was designed from a portion of this cohort (the training set) and tested on the remaining part of the cohort (the test set). Shapley values were used to explain the XGBoost's model features. Model performance was compared with 2 contemporary models designed to predict pancreatic cancer among patients with new-onset diabetes. RESULTS In the test set, the XGBoost model had an area under the curve of 0.80 (0.76 to 0.85) compared with 0.63 and 0.68 for other models. Using cutoffs based on the Youden index, the sensitivity of the XGBoost model was 75%, the specificity was 70%, the accuracy was 70%, the positive predictive value was 1.2%, and the negative predictive value was >99%. The XGBoost model obtained a positive predictive value of at least 2.5% with a sensitivity of 38%. The XGBoost model was the only model that detected at least 50% of patients with cancer one year after the onset of diabetes. All 3 models had similar features that predicted pancreatic cancer, including older age, weight loss, and the rapid destabilization of glucose homeostasis. CONCLUSION Machine learning models isolate a high-risk cohort from those with new-onset diabetes at risk for pancreatic cancer.
Collapse
Affiliation(s)
- Salman Khan
- Department of Medicine, West Virginia University School of Medicine, West Virginia University, Morgantown, WV
- Northeast Ohio Medical University, Rootstown, OH
| | - Bharath Bhushan
- Department of Medicine, West Virginia University School of Medicine, West Virginia University, Morgantown, WV
| |
Collapse
|
43
|
Jacobs MF, Stoffel EM. Genetic and other risk factors for pancreatic ductal adenocarcinoma (PDAC). Fam Cancer 2024; 23:221-232. [PMID: 38573398 DOI: 10.1007/s10689-024-00372-5] [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: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 04/05/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is often diagnosed at an advanced stage, resulting in poor prognosis and low 5-year survival rates. While early evidence suggests increased long-term survival in those with screen-detected resectable cancers, surveillance imaging is currently only recommended for individuals with a lifetime risk of PDAC ≥ 5%. Identification of risk factors for PDAC provides opportunities for early detection, risk reducing interventions, and targeted therapies, thus potentially improving patient outcomes. Here, we summarize modifiable and non-modifiable risk factors for PDAC. We review hereditary cancer syndromes associated with risk for PDAC and their implications for patients and their relatives. In addition, other biologically relevant pathways and environmental and lifestyle risk factors are discussed. Future work may focus on elucidating additional genetic, environmental, and lifestyle risk factors that may modify PDAC risk to continue to identify individuals at increased risk for PDAC who may benefit from surveillance and risk reducing interventions.
Collapse
Affiliation(s)
- Michelle F Jacobs
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Elena M Stoffel
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| |
Collapse
|
44
|
Bogdanski AM, Onnekink AM, Inderson A, Boekestijn B, Bonsing BA, Vasen HFA, van Hooft JE, Boonstra JJ, Mieog JSD, Wasser MNJM, Feshtali S, Potjer TP, Klatte DCF, van Leerdam ME. The Added Value of Blood Glucose Monitoring in High-Risk Individuals Undergoing Pancreatic Cancer Surveillance. Pancreas 2024; 53:e566-e572. [PMID: 38598368 DOI: 10.1097/mpa.0000000000002335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
OBJECTIVES The study aimed to investigate the added value of blood glucose monitoring in high-risk individuals (HRIs) participating in pancreatic cancer surveillance. MATERIALS AND METHODS High-risk individuals with a CDKN2A/p16 germline pathogenic variant participating in pancreatic cancer surveillance were included in this study. Multivariable logistic regression was performed to assess the relationship between new-onset diabetes (NOD) and pancreatic ductal adenocarcinoma (PDAC). To quantify the diagnostic performance of NOD as a marker for PDAC, receiver operating characteristic curve with area under the curve was computed. RESULTS In total, 220 HRIs were included between 2000 and 2019. Median age was 61 (interquartile range. 53-71) years and 62.7% of participants were female. During the study period, 26 (11.8%) HRIs developed NOD, of whom 5 (19.2%) later developed PDAC. The other 23 (82.1%) PDAC cases remained NOD-free. Multivariable analysis showed no statistically significant relationship between NOD and PDAC (odds ratio, 1.21; 95% confidence interval, 0.39-3.78) and 4 of 5 PDAC cases seemed to have NOD within 3 months before diagnosis. Furthermore, NOD did not differentiate between HRIs with and without PDAC (area under the curve, 0.54; 95% confidence interval, 0.46-0.61). CONCLUSIONS In this study, we found no added value for longitudinal glucose monitoring in CDKN2A pathogenic variant carriers participating in an imaging-based pancreatic cancer surveillance program.
Collapse
Affiliation(s)
| | | | - Akin Inderson
- From the Departments of Gastroenterology and Hepatology
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
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
|
46
|
Goggins M. The role of biomarkers in the early detection of pancreatic cancer. Fam Cancer 2024; 23:309-322. [PMID: 38662265 PMCID: PMC11309746 DOI: 10.1007/s10689-024-00381-4] [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: 03/09/2024] [Accepted: 03/19/2024] [Indexed: 04/26/2024]
Abstract
Pancreatic surveillance can detect early-stage pancreatic cancer and achieve long-term survival, but currently involves annual endoscopic ultrasound and MRI/MRCP, and is recommended only for individuals who meet familial/genetic risk criteria. To improve upon current approaches to pancreatic cancer early detection and to expand access, more accurate, inexpensive, and safe biomarkers are needed, but finding them has remained elusive. Newer approaches to early detection, such as using gene tests to personalize biomarker interpretation, and the increasing application of artificial intelligence approaches to integrate complex biomarker data, offer promise that clinically useful biomarkers for early pancreatic cancer detection are on the horizon.
Collapse
Affiliation(s)
- Michael Goggins
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD, 21231, USA.
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
47
|
Satoh T, Nakatani E, Ariyasu H, Kawaguchi S, Ohno K, Itoh H, Hayashi K, Usui T. Pancreatic cancer risk in diabetic patients using the Japanese Regional Insurance Claims. Sci Rep 2024; 14:16958. [PMID: 39043788 PMCID: PMC11266625 DOI: 10.1038/s41598-024-67505-9] [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: 02/13/2024] [Accepted: 07/11/2024] [Indexed: 07/25/2024] Open
Abstract
Pancreatic cancer presents a critical health issue characterized by low survival rates. Identifying risk factors in specific populations, such as those with diabetes, is crucial for early detection and improved outcomes. This study aimed to identify risk factors for pancreatic cancer in diabetic patients using a longitudinal cohort from the Shizuoka Kokuho database, spanning April 2012 to September 2021. Diabetic patients were identified and monitored for the onset of pancreatic cancer. Factors analyzed included age, sex, the Elixhauser comorbidity index, and specific comorbidities. Statistical analyses involved univariate and multivariate Cox proportional hazards regression. The study identified 212,775 as diabetic patients and 1755 developed pancreatic cancer during the period. The annual incidence rate of pancreatic cancer in this group was 166.7 cases per 100,000 person-years. The study identified older age, male sex, a history of liver disease, chronic pancreatitis, and pancreatic cystic lesions as significant risk factors for pancreatic cancer in diabetic patients. The study also highlighted the absence of a significant association between diabetes type or diabetic complications and the onset of pancreatic cancer. These findings may aid in the early diagnosis of pancreatic cancer in diabetic patients and may inform revisions in screening practices in diabetic patients.
Collapse
Affiliation(s)
- Tatsunori Satoh
- Division of Endocrinology, Metabolism and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Shizuoka General Hospital, Shizuoka, Japan
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, 4-27-2 Kitaando, Aoi-Ku, Shizuoka, 420-0881, Japan
| | - Eiji Nakatani
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, 4-27-2 Kitaando, Aoi-Ku, Shizuoka, 420-0881, Japan.
| | - Hiroyuki Ariyasu
- Department of Diabetes and Endocrinology, Shizuoka General Hospital, Shizuoka, Japan
| | - Shinya Kawaguchi
- Department of Gastroenterology, Shizuoka General Hospital, Shizuoka, Japan
| | - Kazuya Ohno
- Department of Gastroenterology, Shizuoka General Hospital, Shizuoka, Japan
| | - Hiroshi Itoh
- Center for Preventive Medicine, Keio University, Tokyo, Japan
| | - Kaori Hayashi
- Division of Endocrinology, Metabolism and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takeshi Usui
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, 4-27-2 Kitaando, Aoi-Ku, Shizuoka, 420-0881, Japan
- Research Support Center, Shizuoka General Hospital, Shizuoka, Japan
| |
Collapse
|
48
|
Aaltonen P, Mustonen H, Puolakkainen P, Haglund C, Peltola K, Carpén O, Lassila R, Seppänen H. Venous thromboembolism incidence and association with overall survival in pancreatic cancer: A Finnish nationwide cohort study. Cancer Med 2024; 13:e70014. [PMID: 39041308 PMCID: PMC11263919 DOI: 10.1002/cam4.70014] [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: 01/23/2024] [Revised: 05/06/2024] [Accepted: 07/04/2024] [Indexed: 07/24/2024] Open
Abstract
INTRODUCTION Pancreatic cancer (PC) is associated with a high risk of venous thromboembolic events (VTEs). We investigated the incidence of VTE before and after the diagnosis of PC and its association with overall survival. METHODS We identified PC patients diagnosed in 2013-2016 from the Finnish Cancer Registry. Data on healthcare visits and death were collected, along with follow-up data through the end of 2020. We compared patients who underwent radical-intent surgery (RIS) to those who underwent palliative treatment (PT) alone. RESULTS We identified 4086 PC patients, of whom 343 (8.4%) underwent RIS and 3743 (91.6%) received PT. VTE incidence within 1 year before a PC diagnosis was higher in the PT (4.2%, n = 156) than in the RIS group (0.6%, n = 2; p < 0.001). The cumulative incidence of VTE at 12 and 24 months after a PC diagnosis was 6% (n = 21) and 9% (n = 31), respectively, within the RIS group, and 8% (n = 286) and 8% (n = 304) within the PT group. In the PT group, a VTE within 1 year before a PC diagnosis was independently associated with a worse survival {hazard ratio, HR 1.9 [95% confidence interval (CI) 1.6-2.2]}. In both groups, VTE after a PC diagnosis was associated with a worse survival [RIS group: HR 2.6 (95%CI 1.8-3.7) vs. PT group: HR 2.2 (95%CI 1.9-2.4)]. CONCLUSIONS A VTE within 1 year before a PC diagnosis more often occurred among PT PC patients than among patients who underwent RIS. VTE might serve as a diagnostic clue to detect PC at an earlier stage.
Collapse
Affiliation(s)
- Panu Aaltonen
- Department of Surgery, Translational Cancer Medicine Research Program, iCAN Digital Precision Cancer Medicine Flagship, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Harri Mustonen
- Department of Surgery, Translational Cancer Medicine Research Program, iCAN Digital Precision Cancer Medicine Flagship, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pauli Puolakkainen
- Department of Surgery, Translational Cancer Medicine Research Program, iCAN Digital Precision Cancer Medicine Flagship, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Caj Haglund
- Department of Surgery, Translational Cancer Medicine Research Program, iCAN Digital Precision Cancer Medicine Flagship, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Katriina Peltola
- Comprehensive Cancer Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Olli Carpén
- Medicum, Research Program in Systems Oncology and HUSLAB, University of Helsinki, Helsinki, Finland
| | - Riitta Lassila
- Department of Hematology, Coagulation Disorders Unit and Research Program Unit in Systems Oncology (ONCOSYS), Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Hanna Seppänen
- Department of Surgery, Translational Cancer Medicine Research Program, iCAN Digital Precision Cancer Medicine Flagship, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
49
|
Lyu J, Jiang M, Zhu Z, Wu H, Kang H, Hao X, Cheng S, Guo H, Shen X, Wu T, Chang J, Wang C. Identification of biomarkers and potential therapeutic targets for pancreatic cancer by proteomic analysis in two prospective cohorts. CELL GENOMICS 2024; 4:100561. [PMID: 38754433 PMCID: PMC11228889 DOI: 10.1016/j.xgen.2024.100561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/12/2023] [Accepted: 04/21/2024] [Indexed: 05/18/2024]
Abstract
Pancreatic cancer (PC) is the deadliest malignancy due to late diagnosis. Aberrant alterations in the blood proteome might serve as biomarkers to facilitate early detection of PC. We designed a nested case-control study of incident PC based on a prospective cohort of 38,295 elderly Chinese participants with ∼5.7 years' follow-up. Forty matched case-control pairs passed the quality controls for the proximity extension assay of 1,463 serum proteins. With a lenient threshold of p < 0.005, we discovered regenerating family member 1A (REG1A), REG1B, tumor necrosis factor (TNF), and phospholipase A2 group IB (PLA2G1B) in association with incident PC, among which the two REG1 proteins were replicated using the UK Biobank Pharma Proteomics Project, with effect sizes increasing steadily as diagnosis time approaches the baseline. Mendelian randomization analysis further supported the potential causal effects of REG1 proteins on PC. Taken together, circulating REG1A and REG1B are promising biomarkers and potential therapeutic targets for the early detection and prevention of PC.
Collapse
Affiliation(s)
- Jingjing Lyu
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minghui Jiang
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziwei Zhu
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongji Wu
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haonan Kang
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjie Hao
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanshan Cheng
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huan Guo
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xia Shen
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
| | - Tangchun Wu
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jiang Chang
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Health Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chaolong Wang
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
50
|
Sapoor S, Nageh M, Shalma NM, Sharaf R, Haroun N, Salama E, Pratama Umar T, Sharma S, Sayad R. Bidirectional relationship between pancreatic cancer and diabetes mellitus: a comprehensive literature review. Ann Med Surg (Lond) 2024; 86:3522-3529. [PMID: 38846873 PMCID: PMC11152885 DOI: 10.1097/ms9.0000000000002036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/30/2024] [Indexed: 06/09/2024] Open
Abstract
Pancreatic cancer (PC) is a fatal malignant disease. It is well known that the relationship between PC and type 2 diabetes mellitus (T2DM) is a complicated bidirectional relationship. The most important factors causing increased risks of pancreatic cancer are hyperglycaemia, hyperinsulinemia, pancreatitis, and dyslipidemia. Genetics and the immune system also play an important role in the relationship between diabetes mellitus and pancreatic cancer. The primary contributors to this association involve insulin resistance and inflammatory processes within the tumour microenvironment. The combination of diabetes and obesity can contribute to PC by inducing hyperinsulinemia and influencing leptin and adiponectin levels. Given the heightened incidence of pancreatic cancer in diabetes patients compared to the general population, early screening for pancreatic cancer is recommended. Diabetes negatively impacts the survival of pancreatic cancer patients. Among patients receiving chemotherapy, it reduced their survival. The implementation of a healthy lifestyle, including weight management, serves as an initial preventive measure to mitigate the risk of disease development. The role of anti-diabetic drugs on survival is controversial; however, metformin may have a positive impact, especially in the early stages of cancer, while insulin therapy increases the risk of PC.
Collapse
Affiliation(s)
| | | | | | - Rana Sharaf
- Faculty of Medicine, Alexandria University, Alexandria
| | - Nooran Haroun
- Faculty of Medicine, Alexandria University, Alexandria
| | - Esraa Salama
- Faculty of Medicine, Alexandria University, Alexandria
| | | | | | - Reem Sayad
- Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|