1
|
Fang Y, Tan C, Zheng Z, Yang J, Tang J, Guo R, Silli EK, Chen Z, Chen J, Ge R, Liu Y, Wen X, Liang J, Zhu Y, Jin Y, Li Q, Wang Y. The function of microRNA related to cancer-associated fibroblasts in pancreatic ductal adenocarcinoma. Biochem Pharmacol 2025; 236:116849. [PMID: 40056941 DOI: 10.1016/j.bcp.2025.116849] [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: 11/11/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignant tumor characterized by a poor prognosis. A prominent feature of PDAC is the rich and dense stroma present in the tumor microenvironment (TME), which significantly hinders drug penetration. Cancer-associated fibroblasts (CAFs), activated fibroblasts originating from various cell sources, including pancreatic stellate cells (PSCs) and mesenchymal stem cells (MSCs), play a critical role in PDAC progression and TME formation. MicroRNAs (miRNAs) are small, single-stranded non-coding RNA molecules that are frequently involved in tumorigenesis and progression, exhibiting either oncolytic or oncogenic activity. Increasing evidence suggests that aberrant expression of miRNAs can mediate interactions between cancer cells and CAFs, thereby providing novel therapeutic targets for PDAC treatment. In this review, we will focus on the potential roles of miRNAs that target CAFs or CAFs-derived exosomes in PDAC progression, highlighting the feasibility of therapeutic strategies aimed at restoring aberrantly expressed miRNAs associated with CAFs, offering new pathways for the clinical management of PDAC.
Collapse
Affiliation(s)
- Yaohui Fang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Chunlu Tan
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenjiang Zheng
- Department of Pancreatic Surgery and General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianchen Yang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jiali Tang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruizhe Guo
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Epiphane K Silli
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Zhe Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jia Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ruyu Ge
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yuquan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xiuqi Wen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jingdan Liang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yunfei Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yutong Jin
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Qian Li
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ying Wang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| |
Collapse
|
2
|
Dreyer SB, Beer P, Hingorani SR, Biankin AV. Improving outcomes of patients with pancreatic cancer. Nat Rev Clin Oncol 2025; 22:439-456. [PMID: 40329051 DOI: 10.1038/s41571-025-01019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 05/08/2025]
Abstract
Research studies aimed at improving the outcomes of patients with pancreatic ductal adenocarcinoma (PDAC) have brought about limited progress, and in clinical practice, the optimized use of surgery, chemotherapy and supportive care have led to modest improvements in survival that have probably reached a plateau. As a result, PDAC is expected to be the second leading cause of cancer-related death in Western societies within a decade. The development of therapeutic advances in PDAC has been challenging owing to a lack of actionable molecular targets, a typically immunosuppressive microenvironment, and a disease course characterized by rapid progression and clinical deterioration. Yet, the progress in our understanding of PDAC and identification of novel therapeutic opportunities over the past few years is leading to a strong sense of optimism in the field. In this Perspective, we address the aforementioned challenges, including biological aspects of PDAC that make this malignancy particularly difficult to treat. We explore specific areas with potential for therapeutic advances, including targeting mutant KRAS, novel strategies to harness the antitumour immune response and approaches to early detection, and propose mechanisms to improve clinical trial design and to overcome various community and institutional barriers to progress.
Collapse
Affiliation(s)
- Stephan B Dreyer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
- Department of Hepatobiliary Surgery, Royal Liverpool University Hospital, Liverpool, UK
| | - Philip Beer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK
- Hull York Medical School, University of York, York, UK
| | - Sunil R Hingorani
- Department of Internal Medicine, Division of Hemotology/Oncology, University of Nebraska Medical Center, Omaha, NE, USA
- Pancreatic Cancer Center of Excellence, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Glasgow, UK.
- West of Scotland Hepato-Biliary and Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK.
| |
Collapse
|
3
|
Qi J, Fang C, Deng C, Shi F, Yao Q. Mesoporous Magnetic Graphene for Serum Metabolic Profiling in Non-Invasive Early Detection and Diagnosis of Pancreatic Ductal Adenocarcinoma. ACS APPLIED MATERIALS & INTERFACES 2025; 17:29995-30005. [PMID: 40355809 DOI: 10.1021/acsami.5c03176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive and lethal cancer, typically diagnosed at advanced stages due to its asymptomatic onset and challenges in early detection. To address the critical need for the early diagnosis of PDAC, we developed a laser desorption/ionization mass spectrometry (LDI-MS) platform based on mesoporous silica-modified magnetic graphene (MG@mSiO2). MG@mSiO2 exhibited exceptional ultraviolet (UV) absorption, efficient ionization, and minimal background interference, enabling high-resolution profiling of serum metabolic fingerprints (SMFs). Based on the extracted SMFs, we constructed a Random Forest (RF) model to classify PDAC patients, high-risk (HR) individuals, and healthy controls (HC), achieving an accuracy of 97.5% in the independent test set. Additionally, a six-metabolite biomarker panel was identified, showing strong diagnostic potential with sensitivity and accuracy exceeding 89.1% for distinguishing HC from PDAC. When coupled with the serological marker carbohydrate antigen 19-9 (CA19-9), the integrated strategy delivered significantly improved diagnostic performance, achieving high accuracy ranging from 95.3% to 100% in distinguishing HR and PDAC patients from HC. Furthermore, metabolic pathway analysis revealed key pathways associated with PDAC progression, providing mechanistic insights into the disease. This work provides a powerful diagnostic tool for PDAC screening, establishing a foundation for early detection and precision medicine in clinical practice.
Collapse
Affiliation(s)
- Jia Qi
- Department of Chemistry, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Caiyun Fang
- Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Chunhui Deng
- Department of Chemistry, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai 200433, China
| | - Fangying Shi
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China
| | - Qunyan Yao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Gastroenterology and Hepatology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, China
| |
Collapse
|
4
|
Lee AA, Twoy A, Sutton A, Fushimi N, Park WG. Suboptimal adherence to surveillance in high-risk individuals for pancreatic cancer at a tertiary care academic center: Lessons from real-world surveillance patterns. Pancreatology 2025:S1424-3903(25)00087-0. [PMID: 40382255 DOI: 10.1016/j.pan.2025.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 05/02/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND/OBJECTIVES Screening for pancreatic ductal adenocarcinoma alters the course of disease among high-risk individuals (HRIs) and is recommended by multiple societies including the International Cancer of the Pancreas Screening Consortium, American Gastroenterological Association, and American Society of Gastrointestinal Endoscopy. However, there are limited analyses on the screening rates and barriers to adherence among HRIs. This study aims to describe real-world longitudinal screening adherence of a HRI surveillance cohort and identify potential barriers to adherence. METHODS Patients followed by Stanford's Benign Pancreas Clinic were identified as HRIs if they met screening criteria per the latest abovementioned screening guidelines, and were included in our study if they underwent at least 1 screening exam. Data on HRIs were obtained retrospectively from our hospital's electronic health record system. Patient and screening characteristics were analyzed by adherence rates. RESULTS 262 HRIs undergoing recommended screening were identified. The mean follow-up time per patient was 4.9 years and the mean successful annual screening rate within the entire cohort was 67%. HRIs in the lowest quartile of adherence were more likely to have more EUS rather than MRI surveillance exams compared to those who were completely adherent (p = 0.01). HRIs who were completely adherent were also an older cohort compared to those with non-complete adherence (p = 0.02) or in the lowest quartile of adherence (p = 0.01). CONCLUSIONS It is difficult to achieve high adherence rates for annual pancreatic cancer screening of HRIs as recommended by the latest guidelines. Age and screening modality may be factors that contribute to adherence difficulty.
Collapse
Affiliation(s)
- Alice A Lee
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA.
| | - Abigail Twoy
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Audrey Sutton
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Naoko Fushimi
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Walter G Park
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| |
Collapse
|
5
|
Bhutani MS, Faraoni EY, Mork ME, McAllister F. Gastric cancer prevention and screening during pancreatic cancer screening in high-risk individuals: an opportunity not to be missed. Gastrointest Endosc 2025; 101:1073-1076. [PMID: 39653170 DOI: 10.1016/j.gie.2024.12.001] [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: 06/18/2024] [Revised: 11/13/2024] [Accepted: 12/02/2024] [Indexed: 01/18/2025]
Affiliation(s)
- Manoop S Bhutani
- Department of Gastroenterology, Hepatology, and Nutrition, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Erika Y Faraoni
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maureen E Mork
- Clinical Cancer Genetics Program, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Florencia McAllister
- Department of Genetics, Clinical Cancer Genetics Program, Department of Gastrointestinal Medical Oncology, Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
6
|
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
|
7
|
Liu Y, Li B, Ke L, Luo T, Wu H, Lin J, Deng Y, Huang X, Xu L, Liu Y, Qi J. Comprehensive Bioinformatics Analyses and Experimental Validation of the Cell Cycle Related Protein SAPCD2 as a New Biomarker and Potential Therapeutic Target in Pancreatic Cancer. J Inflamm Res 2025; 18:2855-2877. [PMID: 40034688 PMCID: PMC11873019 DOI: 10.2147/jir.s501850] [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: 10/22/2024] [Accepted: 02/05/2025] [Indexed: 03/05/2025] Open
Abstract
Purpose Pancreatic adenocarcinoma (PAAD) is a highly aggressive cancer with a poor prognosis, reliable markers are urgently needed for early detection and prognosis evaluation. SAPCD2, a cell cycle related gene, has been implicated in tumorigenesis and proposed as a potential therapeutic target in cancer. However, no comprehensive study has explored its expression and regulation, discussed its role in tumor prognosis and immune modulation, along with therapy response in pan-cancer until now. Methods SAPCD2 expression was analyzed using data from The Cancer Genome Atlas database (TCGA) and Human Protein Atlas (HPA) database. Genetic and epigenetic alterations of SAPCD2 and the immune microenvironment were explored via NCBI, TIMER2 and cBioPortal platforms. Western blot analysis and immunohistochemistry (IHC) were performed to check SAPCD2 protein expression in PAAD cells and tissues. Cell counting kit 8 (CCK8), flow cytometry, and transwell experiments were used to evaluate the role of SAPCD2 in PAAD cell lines. Results Our study found that SAPCD2 is notably upregulated in various cancers, especially early-stage digestive cancers, and is linked to poor survival in most cancers like PAAD and LIHC. Gene amplification and promoter DNA hypomethylation appear to drive this upregulation. Additionally, SAPCD2 expression correlates with tumor mutation burden, microsatellite instability, and immune scores across several cancers. In PAAD, elevated SAPCD2 levels correlated with reduced immune activity, whereas in stomach cancer (STAD), its prognostic impact appeared immune-independent. In PDAC cell lines, SAPCD2 knockdown reduced proliferation and invasion, and caused reduction of G0/G1 phase. PAAD cells with high SAPCD2 expression showed increased sensitivity to DNA-PK, p38α MAPK, and Bcl-2 inhibitors. Conclusion SAPCD2 serves as both a prognostic marker and a potential therapeutic target in PAAD, where its low expression may enhance responsiveness to specific drugs. These findings underscore SAPCD2's dual role in cancer progression and therapy.
Collapse
Affiliation(s)
- Yuting Liu
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
- Scientific Research Center, Guangdong-Hong Kong-Macau University Joint Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Bo Li
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
- Scientific Research Center, Guangdong-Hong Kong-Macau University Joint Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Lingling Ke
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Tingting Luo
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Huixian Wu
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Jiahui Lin
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Yu Deng
- Laboratory Animal Center, Sun Yat-sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Xiuji Huang
- Department of Respiratory and Critical Care Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Liangliang Xu
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, People’s Republic of China
| | - Yuchen Liu
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
- Scientific Research Center, Guangdong-Hong Kong-Macau University Joint Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Jian Qi
- Digestive Diseases Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| |
Collapse
|
8
|
Heller M, Mann DA, Katona BW. Current Approaches of Pancreatic Cancer Surveillance in High-Risk Individuals. J Gastrointest Cancer 2025; 56:61. [PMID: 39932614 PMCID: PMC11814005 DOI: 10.1007/s12029-025-01184-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2025] [Indexed: 02/14/2025]
Abstract
Currently, those recommended to undergo pancreatic cancer (PC) surveillance include appropriately aged individuals at high risk of PC due to an identifiable genetic susceptibility or those without identifiable genetic susceptibility who nonetheless have a strong family history of PC. With increases in identification of individuals at high risk for PC and increased use of PC surveillance in clinical practice, there has been increasing debate about who should undergo surveillance as well as how surveillance should be performed including use of imaging and blood-based testing. Furthermore, there is increasing interest in the outcomes of PC surveillance in high-risk individuals with some studies demonstrating that surveillance leads to downstaging of PC and improvements in survival. In this review, we summarize the current state of PC surveillance in high-risk individuals, providing an overview of the risk factors associated with PC, selection of high-risk individuals for PC surveillance, and the current, but non-uniform, recommendations for performing PC surveillance. Additionally, we review approaches to apply various imaging and blood-based tests to surveillance and the outcomes of PC surveillance.
Collapse
Affiliation(s)
- Melissa Heller
- Division of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Derek A Mann
- Division of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bryson W Katona
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd., 751 South Pavilion, Philadelphia, PA, 19104, USA.
| |
Collapse
|
9
|
Bogdanski AM, Acedo P, Wallace MB, van Leerdam ME, Klatte DCF. Recommendations, evidence and sustainability of screening for pancreatic cancer in high-risk individuals. Best Pract Res Clin Gastroenterol 2025; 74:101974. [PMID: 40210328 DOI: 10.1016/j.bpg.2025.101974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 12/31/2024] [Indexed: 04/12/2025]
Abstract
Pancreatic cancer is a highly lethal malignancy and is predicted to become the second leading cause of cancer-related deaths by 2030. Early detection significantly improves outcomes, but general population screening remains infeasible due to the low prevalence of the disease and lack of specific biomarkers. This review evaluates current recommendations for pancreatic cancer surveillance in high-risk individuals, synthesises evidence from recent studies and explores the sustainability of current imaging-based surveillance programmes. Challenges such as overdiagnosis, economic feasibility and disparities in access highlight the need for targeted, cost-effective strategies. Collaborative initiatives and consortia are needed to advance biomarker research and refine risk stratification. By integrating evidence-based recommendations with sustainable approaches, this review outlines pathways to improve early detection and reduce mortality from pancreatic cancer.
Collapse
Affiliation(s)
- Aleksander M Bogdanski
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, United States; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Pilar Acedo
- Institute for Liver and Digestive Health, Division of Medicine, University College London, London, United Kingdom
| | - Michael B Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, United States
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands; Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Derk C F Klatte
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, United States; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
10
|
Vlăduț C, Steiner C, Löhr M, Gökçe DT, Maisonneuve P, Hank T, Öhlund D, Sund M, Hoogenboom SA. High prevalence of pancreatic steatosis in pancreatic cancer patients: A meta-analysis and systematic review. Pancreatology 2025; 25:98-107. [PMID: 39706752 DOI: 10.1016/j.pan.2024.11.010] [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: 08/29/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 12/23/2024]
Abstract
OBJECTIVE In the last decade there has been increasing interest in defining pancreatic steatosis (PS) and establishing its association with pancreatic ductal adenocarcinoma (PDAC). However, no consensus guidelines have yet been published on the management of PS. In this systematic review and meta-analysis performed according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we investigated the association between PS and PDAC. DESIGN Medical literature between 2007 and 2023 was reviewed for eligible trials investigating the prevalence of PS in patients with PDAC. Eligible studies reporting on PS, assessed via imaging or histology, were included. The primary objective was to determine the association between PDAC and PS by comparing the prevalence of PS in individuals with- and without PDAC. Secondary, an evaluation was conducted to establish whether the method of assessment correlated with the association of PDAC and PS, and the prevalence of PDAC in individuals with PS. Measures of effect size were determined using odds ratios (ORs) and corresponding 95 % confidence intervals (95 % CI). RESULTS The systematic review identified a total of 23 studies, of which seventeen studies examined PS prevalence among PDAC patients and were included in the meta-analysis. Overall, the pooled prevalence of PS in patients with PDAC was 53.6 % (95 % CI 40.9-66.2). No significant difference in PS prevalence was observed across various diagnostic methods or geographical regions. Overall, the pooled OR for PS in patients with PDAC compared to controls was 3.23 (95 % CI 1.86-5.60). CONCLUSIONS PDAC patients have a high prevalence of PS, and they are significantly more likely to have PS compared to controls. These findings emphasize the need to prioritize a standardized approach to the diagnosis, follow-up, and treatment of PS, with future studies focusing on identifying patients who would benefit from PDAC surveillance programs.
Collapse
Affiliation(s)
- Cătălina Vlăduț
- Department of Gastroenterology, "Prof Dr Agrippa Ionescu" Clinical Emergency Hospital, 011356 Bucharest, Romania; Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania.
| | | | - Matthias Löhr
- Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden.
| | - Dilara Turan Gökçe
- Department of Gastroenterology, Ankara Bilkent City Hospital, Ankara, Turkey.
| | - Patrick Maisonneuve
- Division of Epidemiology and Biostatistics, IEO European Institute of Oncology IRCCS, Milan, Italy.
| | - Thomas Hank
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany.
| | - Daniel Öhlund
- Department of Diagnostics and Intervention (oncology) and Wallenberg Centre of Molecular Medicine (WCMM), Umeå University, Umeå, Sweden.
| | - Malin Sund
- Department of Diagnostics and Intervention (surgery), Umeå University, Umeå, Sweden; Department of Surgery, University of Helsinki and Helsinki, University Hospital, Helsinki, Finland.
| | - Sanne A Hoogenboom
- Department of Gastroenterology, HagaZiekenhuis Hospital, The Hague, Netherlands.
| |
Collapse
|
11
|
Athanasiou A, Kureshi N, Wittig A, Sterner M, Huber R, Palma NA, King T, Schiess R. Biomarker Discovery for Early Detection of Pancreatic Ductal Adenocarcinoma (PDAC) Using Multiplex Proteomics Technology. J Proteome Res 2025; 24:315-322. [PMID: 39699878 PMCID: PMC11705213 DOI: 10.1021/acs.jproteome.4c00752] [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] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/27/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Early detection of pancreatic ductal adenocarcinoma (PDAC) can improve survival but is hampered by the absence of early disease symptoms. Imaging remains key for surveillance but is cumbersome and may lack sensitivity to detect small tumors. CA19-9, the only FDA-approved blood biomarker for PDAC, is insufficiently sensitive and specific to be recommended for surveillance. We aimed to discover a blood-based protein signature to improve PDAC detection in our main target population consisting of stage I or II PDAC patients (n = 75) and various controls including healthy controls (n = 50), individuals at high risk (genetic and familial) for PDAC (n = 47), or those under surveillance for an intraductal papillary mucinous neoplasm (n = 36). Roughly 3000 proteins were measured using Olink multiplex technology and conventional immunoassays. Machine learning combined biomarker candidates into 4- to 6-plex signatures. These signatures significantly (p < 0.001) outperformed CA19-9 with 84% sensitivity at 95% specificity, compared to CA19-9's sensitivity of 53% in the target population. Exploratory analysis was performed in new-onset diabetes (n = 81) and chronic pancreatitis (n = 50) patients. In conclusion, 41 promising biomarker candidates across multiple signatures were identified using proteomics technology and will be further tested in an independent cohort.
Collapse
Affiliation(s)
| | - Natasha Kureshi
- Immunovia
Inc., 26 Forest Street,
Suite 110, Marlborough, Massachusetts 01752, United States
| | - Anja Wittig
- Proteomedix
AG, Wagistrasse 23, CH-8952 Schlieren, Switzerland
| | - Maria Sterner
- Immunovia
AB, Medicon Village,
Scheelevägen 8, SE-223 63 Lund, Sweden
| | - Ramy Huber
- Proteomedix
AG, Wagistrasse 23, CH-8952 Schlieren, Switzerland
| | - Norma A. Palma
- Immunovia
Inc., 26 Forest Street,
Suite 110, Marlborough, Massachusetts 01752, United States
| | - Thomas King
- Immunovia
Inc., 26 Forest Street,
Suite 110, Marlborough, Massachusetts 01752, United States
| | - Ralph Schiess
- Proteomedix
AG, Wagistrasse 23, CH-8952 Schlieren, Switzerland
| |
Collapse
|
12
|
Corallo C, Al-Adhami AS, Jamieson N, Valle J, Radhakrishna G, Moir J, Albazaz R. An update on pancreatic cancer imaging, staging, and use of the PACT-UK radiology template pre- and post-neoadjuvant treatment. Br J Radiol 2025; 98:13-26. [PMID: 39460945 DOI: 10.1093/bjr/tqae217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 10/01/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma continues to have a poor prognosis, although recent advances in neoadjuvant treatments (NATs) have provided some hope. Imaging assessment of suspected tumours can be challenging and requires a specific approach, with pancreas protocol CT being the primary imaging modality for staging with other modalities used as problem-solving tools to facilitate appropriate management. Imaging assessment post NAT can be particularly difficult due to a current lack of robust radiological criteria to predict response and differentiate treatment induced fibrosis/inflammation from residual tumour. This review aims to provide an update of pancreatic ductal adenocarcinoma with particular focus on three points: tumour staging pre- and post-NAT including vascular assessment, structured reporting with introduction of the PAncreatic Cancer reporting Template-UK (PACT-UK) radiology template, and the potential future role of artificial intelligence in the diagnosis and staging of pancreatic cancer.
Collapse
Affiliation(s)
- Carmelo Corallo
- Department of Radiology, St James's University Hospital, Leeds LS9 7TF, United Kingdom
| | - Abdullah S Al-Adhami
- Department of Radiology, Glasgow Royal Infirmary, Glasgow G31 2ER, United Kingdom
| | - Nigel Jamieson
- HPB Unit, Glasgow Royal Infirmary, Glasgow G31 2ER, United Kingdom
| | - Juan Valle
- Division of Cancer Sciences, University of Manchester, Manchester M20 4GJ, United Kingdom
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4 BX, United Kingdom
| | | | - John Moir
- HPB Unit, Freeman Hospital, Newcastle Upon Tyne NE7 7DN, United Kingdom
| | - Raneem Albazaz
- Department of Radiology, St James's University Hospital, Leeds LS9 7TF, United Kingdom
- University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
13
|
Di Y, Song J, Sun Z, Wang Y, Meng L. Nonsurgical pancreatic cancer: the role of radiotherapy in prolonging survival - a retrospective cohort study in the SEER database. Int J Surg 2025; 111:818-827. [PMID: 38920320 PMCID: PMC11745595 DOI: 10.1097/js9.0000000000001885] [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: 02/21/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Limited research has compared external beam radiotherapy (RT) to non-RT in patients with nonsurgical locally advanced pancreatic cancer (LAPC). Therefore, this study investigates the impact of RT on overall survival (OS) in patients with nonsurgical LAPC in a real-world context. METHODS The authors conducted an analysis of patients with nonsurgical LAPC using data from the Surveillance, Epidemiology, and End Results (SEER) database. This analysis involved the utilization of Kaplan-Meier survival curves and multivariable Cox regression analyses. RESULTS A total of 5413 individuals with nonsurgical LAPC were included in this analysis. Among them, 2320 (42.9%) received RT, while 3093 (57.1%) underwent non-RT treatment. The median OS was 12.0 months for the RT group and 9.0 months for the non-RT group, with a statistically significant difference ( P <0.001). Multivariate analysis revealed that RT had a statistically significant impact on OS (HR, 0.86; 95% CI: 0.81-0.91; P <0.001). Propensity score matching analysis confirmed a statistically significant association of RT with improved OS (HR, 0.84, 95% CI: 0.79-0.90; P <0.001). These results remained consistent after conducting sensitivity analyses, subgroup analyses, and propensity score matching. CONCLUSION The study findings suggest that RT could be advantageous for patients with nonsurgical LAPC. Further investigations are warranted to explore the relationship between RT and OS.
Collapse
Affiliation(s)
- Yupeng Di
- Department of Radiation Oncology, Air Force Medical Center, PLA, Beijing, People's Republic of China
| | - Jiazhao Song
- Department of Radiation Oncology, Air Force Medical Center, PLA, Beijing, People's Republic of China
| | - Zhijia Sun
- Department of Radiation Oncology, Air Force Medical Center, PLA, Beijing, People's Republic of China
| | - Yingjie Wang
- Department of Radiation Oncology, Air Force Medical Center, PLA, Beijing, People's Republic of China
| | - Lingling Meng
- Department of Radiation Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
14
|
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
|
15
|
Maurer E, Lehman B, Matthäi E, Denzer U, Figiel J, Jesinghaus M, Slater EP, Stefenelli U, Gress TM, Bartsch DK. Pancreatic cancer screening is effective in individuals at risk with predisposing germline gene variants, but not in gene variant-negative familial pancreatic cancer families. United European Gastroenterol J 2024; 12:1211-1221. [PMID: 39031472 PMCID: PMC11578844 DOI: 10.1002/ueg2.12631] [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: 03/17/2024] [Accepted: 06/20/2024] [Indexed: 07/22/2024] Open
Abstract
OBJECTIVE To evaluate the diagnostic yield of pancreatic cancer screening in individuals at risk (IAR) from familial pancreatic cancer (FPC) families with respect to the presence or absence of pathogenic germline variants predisposing to pancreatic adenocarcinoma (PDAC). DESIGN In a 20 years period, IAR from FPC families were enrolled in a prospective screening program of the national case collection for FPC of Germany, including magnet resonance imaging (MRI) and endoscopic ultrasound (EUS). The diagnostic yield was analyzed regarding significant pancreatic lesions such as PDAC, high-grade pancreatic-intraepithelial-neoplasia (PanIN3) and intraductal-papillary-mucinous-neoplasia (IPMN) with high-grade dysplasia. Screening results were compared between carriers of pathogenic variants and variant-negative IAR. RESULTS 337 IAR, including 74 (22%) variant-carriers and 263 IAR of variant-negative FPC families (mean age 49; standard deviation [SD] + 8.9) were followed 64 (SD + 55) months. IAR underwent 5.1 (SD + 3.9) screening visits with 1733 MRI (5.1,SD + 3.9 per IAR) and 728 EUS (2.2,SD + 1.7 per IAR). In 12 (4%) cases, significant pancreatic lesions were detected, including 4 PDAC, 3 PanIN3 and 5 high-grade IPMN. Three of 4 IAR with PDAC died after a mean of 27 months postoperatively, and one IAR is alive without evidence of disease after 31 months. The diagnostic yield for significant lesions was 13.5% (10/74) for variant carriers compared to 0.8% (2/263) for IAR of variant-negative FPC families (p < 0.001). Logistic regression analysis revealed that a negative variant status was almost always accompanied by the absence of a significant lesion over time with a negative predictive value of 99.2% (95% CI 97.3%-99.9%). CONCLUSION The diagnostic yield seems to justify PDAC screening in IAR of FPC-families with pathogenic germline variants in PDAC predisposing genes, not in IAR of variant-negative families.
Collapse
Affiliation(s)
- Elisabeth Maurer
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| | - Bettina Lehman
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| | - Elvira Matthäi
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| | - Ulrike Denzer
- Department of GastroenterologyEndocrinologyMetabolism and InfectiologyPhilipps University MarburgMarburgGermany
| | - Jens Figiel
- Department of Diagnostic and Interventional RadiologyPhilipps University MarburgMarburgGermany
| | | | - Emily P. Slater
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| | | | - Thomas M. Gress
- Department of GastroenterologyEndocrinologyMetabolism and InfectiologyPhilipps University MarburgMarburgGermany
| | - Detlef K. Bartsch
- Department of Visceral‐Thoracic‐ and Vascular SurgeryPhilipps University MarburgMarburgGermany
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Ahmed TM, Kawamoto S, Lopez-Ramirez F, Yasrab M, Hruban RH, Fishman EK, Chu LC. Early detection of pancreatic cancer in the era of precision medicine. Abdom Radiol (NY) 2024; 49:3559-3573. [PMID: 38761272 DOI: 10.1007/s00261-024-04358-w] [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: 03/31/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/20/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related mortality and it is often diagnosed at advanced stages due to non-specific clinical presentation. Disease detection at localized disease stage followed by surgical resection remains the only potentially curative treatment. In this era of precision medicine, a multifaceted approach to early detection of PDAC includes targeted screening in high-risk populations, serum biomarkers and "liquid biopsies", and artificial intelligence augmented tumor detection from radiologic examinations. In this review, we will review these emerging techniques in the early detection of PDAC.
Collapse
Affiliation(s)
- Taha M Ahmed
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Felipe Lopez-Ramirez
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Mohammad Yasrab
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Ralph H Hruban
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Linda C Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA.
| |
Collapse
|
19
|
Klatte DCF, Onnekink AM, Hinnen C, van Doorn R, Potjer TP, van Leerdam ME, Bleiker EMA. Psychosocial issues of individuals undergoing surveillance for increased risk of melanoma and pancreatic cancer due to a germline CDKN2A variant: A focus group study. J Genet Couns 2024; 33:1059-1069. [PMID: 37876362 DOI: 10.1002/jgc4.1820] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 10/03/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
Individuals with a germline CDKN2A pathogenic variant (PV) are at high risk of developing melanoma and pancreatic cancer and are therefore offered surveillance. The potential advantages and disadvantages associated with genetic testing and surveillance are discussed during medical counseling, although little is known about the associated psychosocial factors that are relevant to this population. This study sought to provide a qualitative exploration of psychosocial factors related to genetic testing and participation in skin and pancreatic surveillance in (potential) carriers of a CDKN2A PV. Fifteen individuals-both at-risk individuals and confirmed variant carriers-participated in one of the three online focus groups. Pre-defined discussion topics, including genetic testing, cancer surveillance, influence on lifestyle and family planning, were discussed. Patients reported that important reasons to engage in genetic testing included the possibility to participate in surveillance to gain control over their cancer risk and to get clarification on the potential carrier status of their children. We observed considerable differences in risk perception and experienced burden of surveillance. Knowledge of the PV has had a positive influence on lifestyle factors and altered attitudes toward life in some. Most participants were not aware of preimplantation genetic testing. This focus group study provided insight into a variety of psychosocial themes related to (potential) carriership of a CDKN2A PV. Future efforts should focus on identifying those who may benefit from additional psychosocial support, development of a centralized source of information, and assessing the knowledge, needs, and timing of counseling for family planning.
Collapse
Affiliation(s)
- Derk C F Klatte
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anke M Onnekink
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Chris Hinnen
- Department of Medical Psychology, Leiden University Medical Center, Leiden, The Netherlands
| | - Remco van Doorn
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas P Potjer
- Department of Clinical Genetics, 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, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Eveline M A Bleiker
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Family Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Kane WJ, Haden KR, Martin EN, Shami VM, Wang AY, Strand DS, Adair SJ, Nagdas S, Tsung A, Zaydfudim VM, Adams RB, Bauer TW. Survival benefit associated with screening of patients at elevated risk for pancreatic cancer. J Surg Oncol 2024; 130:485-492. [PMID: 39016067 DOI: 10.1002/jso.27784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 06/30/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND & OBJECTIVES: Screening for pancreatic cancer is recommended for individuals with a strong family history, certain genetic syndromes, or a neoplastic cyst of the pancreas. However, limited data supports a survival benefit attributable to screening these higher-risk individuals. METHODS All patients enrolled in screening at a High-Risk Pancreatic Cancer Clinic (HRC) from July 2013 to June 2020 were identified from a prospectively maintained institutional database and compared to patients evaluated at a Surgical Oncology Clinic (SOC) at the same institution during the same period. Clinical outcomes of patients selected for surgical resection, particularly clinicopathologic stage and overall survival, were compared. RESULTS Among 826 HRC patients followed for a median (IQR) of 2.3 (0.8-4.2) years, 128 were selected for surgical resection and compared to 402 SOC patients selected for resection. Overall survival was significantly longer among HRC patients (median survival: not reached vs. 2.6 years, p < 0.001). Among 31 HRC and 217 SOC patients with a diagnosis of pancreatic ductal adenocarcinoma (PDAC), the majority of HRC patients were diagnosed with stage 0 disease (carcinoma in situ), while the majority of SOC patients were diagnosed with stage II disease (p < 0.001). Overall survival after resection of invasive PDAC was also significantly longer among HRC patients compared to SOC patients (median survival 5.5 vs. 1.6 years, p = 0.002). CONCLUSION Patients at increased risk for PDAC and followed with guideline-based screening exhibited downstaging of disease and improved survival from PDAC in comparison to patients who were not screened.
Collapse
Affiliation(s)
- William J Kane
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Kathleen R Haden
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Elizabeth N Martin
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Vanessa M Shami
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia, USA
| | - Andrew Y Wang
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel S Strand
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Virginia, Charlottesville, Virginia, USA
| | - Sara J Adair
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Sarbajeet Nagdas
- School of Medicine, University of Virginia, Charlottesville, University of Virginia, USA
| | - Allan Tsung
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Victor M Zaydfudim
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Reid B Adams
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Todd W Bauer
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
21
|
Shah Y, Dahiya DS, Tiwari A, Kumar H, Gangwani MK, Ali H, Hayat U, Alsakarneh S, Singh S, Malik S, Sohail AH, Chandan S, Ali MA, Inamdar S. Advancements in Early Detection and Screening Strategies for Pancreatic Cancer: From Genetic Susceptibility to Novel Biomarkers. J Clin Med 2024; 13:4706. [PMID: 39200847 PMCID: PMC11355237 DOI: 10.3390/jcm13164706] [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: 07/25/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
Pancreatic cancer is a rare but lethal cancer due to its biologically aggressive nature, advanced stage at the time of diagnosis, and poor response to oncologic therapies. The risk of pancreatic cancer is significantly higher to 5% in certain high-risk individuals with inherited genetic susceptibility. Screening for pancreatic cancer in these individuals from high-risk groups can help with the early detection of pancreatic cancer as well as the detection of precursor lesions leading to early surgical resection and improved overall outcomes. The advancements in radiological imaging as well as advanced endoscopic procedures has made a significant impact on the early diagnosis, surveillance, and staging of pancreatic cancer. There is also a significant advancement in the development of biomarkers for the early detection of pancreatic cancer, which has also led to the development of liquid biopsy, allowing for microRNA detection in serum and circulating tumor cells. Various societies and organizations have provided guidelines for pancreatic cancer screening and surveillance in high-risk individuals. In this review, we aim to discuss the hereditary risk factors for developing pancreatic cancer, summarize the screening recommendations by different societies, and discuss the development of novel biomarkers and areas for future research in pancreatic cancer screening for high-risk individuals.
Collapse
Affiliation(s)
- Yash Shah
- Department of Internal Medicine, Trinity Health Oakland/Wayne State University, Pontiac, MI 48341, USA
| | - Dushyant Singh Dahiya
- Division of Gastroenterology, Hepatology & Motility, The University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | - Angad Tiwari
- Department of Internal Medicine, Maharani Laxmi Bai Medical College, Jhansi 284001, Uttar Pradesh, India
| | - Harendra Kumar
- Department of Internal Medicine, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Manesh Kumar Gangwani
- Department of Gastroenterology and Hepatology, University of Arkansas For Medical Sciences, Little Rock, AR 72205, USA
| | - Hassam Ali
- Division of Gastroenterology, Hepatology & Nutrition, East Carolina University/Brody School of Medicine, Greenville, NC 27834, USA
| | - Umar Hayat
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes Barre, PA 18711, USA
| | - Saqr Alsakarneh
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Sahib Singh
- Department of Internal Medicine, Sinai Hospital, Baltimore, MD 21215, USA
| | - Sheza Malik
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY 14621, USA
| | - Amir H. Sohail
- Department of Surgery, University of New Mexico, Albuquerque, NM 87131, USA
| | - Saurabh Chandan
- Center for Interventional Endoscopy (CIE), Advent Health, Orlando, FL 32803, USA
| | - Meer A. Ali
- Department of Gastroenterology and Hepatology, University of Arkansas For Medical Sciences, Little Rock, AR 72205, USA
| | - Sumant Inamdar
- Department of Gastroenterology and Hepatology, University of Arkansas For Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
22
|
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: 5] [Impact Index Per Article: 5.0] [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
|
23
|
Matsubayashi H, Morizane C. Familial and hereditary pancreatic cancer in Japan. Fam Cancer 2024; 23:365-372. [PMID: 38733422 DOI: 10.1007/s10689-024-00395-y] [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: 11/29/2023] [Accepted: 04/19/2024] [Indexed: 05/13/2024]
Abstract
As in Western countries, familial pancreatic cancer accounts for 5-7% of pancreatic cancer (PC) in Japan. Opportunities for diagnosing hereditary pancreatic cancer (HPC) are increasing owing to the coverage of companion diagnostics and cancer genomic profiling by national health insurance in patients with unresectable or recurrent PC refractory to standard chemotherapies. HPC is recognized in 7% of PCs and 15% of familial pancreatic cancer, including germline variants of BRCA1/2, ATM, PALB2, APC, and mismatch repair genes. Individuals with 5-fold or greater inherited risks of PC are recommended to undergo pancreatic surveillance according to Japanese guidelines. The imaging modalities for this surveillance include endoscopic ultrasound, magnetic resonance cholangiopancreatography, abdominal ultrasound, and enhanced computed tomography. Currently, a nationwide prospective surveillance study is ongoing in Japan. Platinum-based chemotherapy is an effective pancreatic cancer treatment in patients with variants of homologous recombination repair genes (BRCA1/2 and PALB2); however, the use of platinum regimens solely based on familial/personal cancer history remains controversial. The efficacy of olaparib maintenance therapy, as confirmed by the POLO study, has significantly impacted the clinical treatment of advanced PC patients in Japan. Since the initiation of precision cancer medicine in 2019, genetic medicine for PC patients has expanded in Japan.
Collapse
Affiliation(s)
- Hiroyuki Matsubayashi
- Division of Genetic Medicine Promotion and Endoscopy, Shizuoka Cancer Center, Shimonagakubo, Nagaizumi, Suntogun, Shizuoka, 411-8777, Japan.
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| |
Collapse
|
24
|
Vasen HF, Canto MI, Goggins M. Twenty-five years of surveillance for familial and hereditary pancreatic ductal adenocarcinoma: Historical perspectives and introduction to the special issue. Fam Cancer 2024; 23:209-215. [PMID: 38844715 PMCID: PMC11255030 DOI: 10.1007/s10689-024-00404-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 07/18/2024]
Abstract
In the 1990s, as prevention became a central strategy in the battle against cancer and the molecular genetics revolution uncovered the genetic basis of numerous hereditary cancer syndromes, there were no options available for patients at increased risk of developing pancreatic cancer. When surveillance efforts for those at familial and hereditary risk of pancreatic cancer emerged in the late 1990s, it was uncertain if early detection was achievable.In this introduction to the special issue, we offer an overview of the history of surveillance for pancreatic cancer, including the first reports of familial pancreatic cancer in the medical literature, the initial results of surveillance in the United States and the initiation of surveillance programs for hereditary pancreatic cancer in the Netherlands.This special issue features a collection of 18 articles written by prominent experts in the field, focusing specifically on refining surveillance methodologies with the primary objective of improving care of high-risk individuals. Several reviews in this collection highlight improved survival rates associated with pancreas surveillance, underlying the potential of early detection and improved management in the continuing fight against pancreatic cancer.
Collapse
Affiliation(s)
- Hans Fa Vasen
- Department of Gastroenterology & Hepatology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Marcia Irene Canto
- Department of Medicine, Division of Gastroenterology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Michael Goggins
- Department of Medicine, Division of Gastroenterology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Department of Pathology, and Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| |
Collapse
|
25
|
Cristina-Marianini-Rios, Sanchez MEC, de Paredes AGG, Rodríguez M, Barreto E, López JV, Fuentes R, Beltrán MM, Sanjuanbenito A, Lobo E, Caminoa A, Ruz-Caracuel I, Durán SL, Olcina JRF, Blázquez J, Sequeros EV, Carrato A, Ávila JCM, Earl J. The best linear unbiased prediction (BLUP) method as a tool to estimate the lifetime risk of pancreatic ductal adenocarcinoma in high-risk individuals with no known pathogenic germline variants. Fam Cancer 2024; 23:233-246. [PMID: 38780705 PMCID: PMC11254992 DOI: 10.1007/s10689-024-00397-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related death in the Western world. The number of diagnosed cases and the mortality rate are almost equal as the majority of patients present with advanced disease at diagnosis. Between 4 and 10% of pancreatic cancer cases have an apparent hereditary background, known as hereditary pancreatic cancer (HPC) and familial pancreatic cancer (FPC), when the genetic basis is unknown. Surveillance of high-risk individuals (HRI) from these families by imaging aims to detect PDAC at an early stage to improve prognosis. However, the genetic basis is unknown in the majority of HRIs, with only around 10-13% of families carrying known pathogenic germline mutations. The aim of this study was to assess an individual's genetic cancer risk based on sex and personal and family history of cancer. The Best Linear Unbiased Prediction (BLUP) methodology was used to estimate an individual's predicted risk of developing cancer during their lifetime. The model uses different demographic factors in order to estimate heritability. A reliable estimation of heritability for pancreatic cancer of 0.27 on the liability scale, and 0.07 at the observed data scale as obtained, which is different from zero, indicating a polygenic inheritance pattern of PDAC. BLUP was able to correctly discriminate PDAC cases from healthy individuals and those with other cancer types. Thus, providing an additional tool to assess PDAC risk HRI with an assumed genetic predisposition in the absence of known pathogenic germline mutations.
Collapse
Affiliation(s)
- Cristina-Marianini-Rios
- Department of Agricultural Economics, Statistics and Business Management, Universidad Politécnica de Madrid, Madrid, Spain
| | - María E Castillo Sanchez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
| | - Ana García García de Paredes
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercedes Rodríguez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- University of Alcalá, Madrid, Spain
| | - Emma Barreto
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- University of Alcalá, Madrid, Spain
| | - Jorge Villalón López
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
| | - Raquel Fuentes
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, 28034, Spain
| | | | - Alfonso Sanjuanbenito
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- Pancreatic and Biliopancreatic Surgery Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Eduardo Lobo
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Pancreatic and Biliopancreatic Surgery Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alejandra Caminoa
- Department of Pathology, Hospital Universitario Ramón y Cajal, Madrid, 28034, Spain
| | - Ignacio Ruz-Caracuel
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- Department of Pathology, Hospital Universitario Ramón y Cajal, Madrid, 28034, Spain
| | - Sergio López Durán
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - José Ramón Foruny Olcina
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Javier Blázquez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Radiology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Enrique Vázquez Sequeros
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
| | - Alfredo Carrato
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain
- University of Alcalá, Madrid, Spain
- Pancreatic Cancer Europe, Brussels, Belgium
| | - Jose Carlos Martínez Ávila
- Department of Agricultural Economics, Statistics and Business Management, Universidad Politécnica de Madrid, Madrid, Spain.
| | - Julie Earl
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, Madrid, 28034, Spain.
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, Madrid, 28029, Spain.
| |
Collapse
|
26
|
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
|
27
|
Blackford AL, Canto MI, Dbouk M, Hruban RH, Katona BW, Chak A, Brand RE, Syngal S, Farrell J, Kastrinos F, Stoffel EM, Rustgi A, Klein AP, Kamel I, Fishman EK, He J, Burkhart R, Shin EJ, Lennon AM, Goggins M. Pancreatic Cancer Surveillance and Survival of High-Risk Individuals. JAMA Oncol 2024; 10:1087-1096. [PMID: 38959011 PMCID: PMC11223057 DOI: 10.1001/jamaoncol.2024.1930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/05/2024] [Indexed: 07/04/2024]
Abstract
Importance Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease with increasing incidence. The majority of PDACs are incurable at presentation, but population-based screening is not recommended. Surveillance of high-risk individuals for PDAC may lead to early detection, but the survival benefit is unproven. Objective To compare the survival of patients with surveillance-detected PDAC with US national data. Design, Setting, and Participants This comparative cohort study was conducted in multiple US academic medical centers participating in the Cancer of the Pancreas Screening program, which screens high-risk individuals with a familial or genetic predisposition for PDAC. The comparison cohort comprised patients with PDAC matched for age, sex, and year of diagnosis from the Surveillance, Epidemiology, and End Results (SEER) program. The Cancer of the Pancreas Screening program originated in 1998, and data collection was done through 2021. The data analysis was performed from April 29, 2022, through April 10, 2023. Exposures Endoscopic ultrasonography or magnetic resonance imaging performed annually and standard-of-care surgical and/or oncologic treatment. Main Outcomes and Measures Stage of PDAC at diagnosis, overall survival (OS), and PDAC mortality were compared using descriptive statistics and conditional logistic regression, Cox proportional hazards regression, and competing risk regression models. Sensitivity analyses and adjustment for lead-time bias were also conducted. Results A total of 26 high-risk individuals (mean [SD] age at diagnosis, 65.8 [9.5] years; 15 female [57.7%]) with PDAC were compared with 1504 SEER control patients with PDAC (mean [SD] age at diagnosis, 66.8 [7.9] years; 771 female [51.3%]). The median primary tumor diameter of the 26 high-risk individuals was smaller than in the control patients (2.5 [range, 0.6-5.0] vs 3.6 [range, 0.2-8.0] cm, respectively; P < .001). The high-risk individuals were more likely to be diagnosed with a lower stage (stage I, 10 [38.5%]; stage II, 8 [30.8%]) than matched control patients (stage I, 155 [10.3%]; stage II, 377 [25.1%]; P < .001). The PDAC mortality rate at 5 years was lower for high-risk individuals than control patients (43% vs 86%; hazard ratio, 3.58; 95% CI, 2.01-6.39; P < .001), and high-risk individuals lived longer than matched control patients (median OS, 61.7 [range, 1.9-147.3] vs 8.0 [range, 1.0-131.0] months; 5-year OS rate, 50% [95% CI, 32%-80%] vs 9% [95% CI, 7%-11%]). Conclusions and Relevance These findings suggest that surveillance of high-risk individuals may lead to detection of smaller, lower-stage PDACs and improved survival.
Collapse
Affiliation(s)
- Amanda L. Blackford
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Marcia Irene Canto
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Mohamad Dbouk
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Ralph H. Hruban
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Bryson W. Katona
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Amitabh Chak
- Division of Gastroenterology and Liver Disease, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, Ohio
| | - Randall E. Brand
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center, Pennsylvania
| | - Sapna Syngal
- Cancer Genetics and Prevention, Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Gastroenterology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - James Farrell
- Yale Center for Pancreatic Disease, Section of Digestive Disease, Yale University, New Haven, Connecticut
| | - Fay Kastrinos
- Division of Digestive and Liver Diseases, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Elena M. Stoffel
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Anil Rustgi
- Division of Digestive and Liver Diseases, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Alison P. Klein
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Ihab Kamel
- Department of Radiology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Elliot K. Fishman
- Department of Radiology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Jin He
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Richard Burkhart
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Eun Ji Shin
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Anne Marie Lennon
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Radiology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Michael Goggins
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| |
Collapse
|
28
|
Maurer E, Bartsch DK. Surgical aspects related to hereditary pancreatic cancer. Fam Cancer 2024; 23:341-350. [PMID: 38662263 PMCID: PMC11254980 DOI: 10.1007/s10689-024-00384-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/29/2024] [Indexed: 04/26/2024]
Abstract
The goal of surveillance programs for individuals at risk (IAR) from familial pancreatic cancer (FPC) families or families with other inherited tumor syndromes predisposing to the development of pancreatic adenocarcinoma (PDAC), such as hereditary pancreatitis or Peutz-Jeghers syndrome, is the dectection and consecutive curative resection of early PDAC or even better its high-grade precursor lesions. Although the indication for surgery is quite established, the extent of surgery is not well defined due to the lack of evidence-based data. In addition, multiple factors have to be taken into account to determine an optimal personalized surgical strategy. This holds especially true since pancreatic surgery is associated with a relatively high morbidity and might impair the quality of life significantly. In this article the surgical aspects in the setting of hereditary PDAC are discussed.
Collapse
Affiliation(s)
- Elisabeth Maurer
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University Marburg, 35043, Baldingerstrasse, Marburg, Germany.
| | - Detlef K Bartsch
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University Marburg, 35043, Baldingerstrasse, Marburg, Germany
| |
Collapse
|
29
|
Boekestijn B, Feshtali S, Vasen H, van Leerdam ME, Bonsing BA, Mieog JSD, Wasser MN. Screening for pancreatic cancer in high-risk individuals using MRI: optimization of scan techniques to detect small lesions. Fam Cancer 2024; 23:295-308. [PMID: 38733421 PMCID: PMC11254973 DOI: 10.1007/s10689-024-00394-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
Pancreatic cancer has a dismal prognosis in the general population. However, early detection and treatment of disease in high-risk individuals can improve survival, as patients with localized disease and especially patients with lesions smaller than 10 mm show greatly improved 5-year survival rates. To achieve early detection through MRI surveillance programs, optimization of imaging is required. Advances in MRI technologies in both hardware and software over the years have enabled reliable detection of pancreatic cancer at a small size and early stage. Standardization of dedicated imaging protocols for the pancreas are still lacking. In this review we discuss state of the art scan techniques, sequences, reduction of artifacts and imaging strategies that enable early detection of lesions. Furthermore, we present the imaging features of small pancreatic cancers from a large cohort of high-risk individuals. Refinement of MRI techniques, increased scan quality and the use of artificial intelligence may further improve early detection and the prognosis of pancreatic cancer in a screening setting.
Collapse
Affiliation(s)
- Bas Boekestijn
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Shirin Feshtali
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans Vasen
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin N Wasser
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
30
|
Overbeek KA, Cahen DL, Bruno MJ. The role of endoscopic ultrasound in the detection of pancreatic lesions in high-risk individuals. Fam Cancer 2024; 23:279-293. [PMID: 38573399 PMCID: PMC11255057 DOI: 10.1007/s10689-024-00380-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/17/2024] [Indexed: 04/05/2024]
Abstract
Individuals at high risk of developing pancreatic ductal adenocarcinoma are eligible for surveillance within research programs. These programs employ periodic imaging in the form of magnetic resonance imaging/magnetic resonance cholangiopancreatography or endoscopic ultrasound for the detection of early cancer or high-grade precursor lesions. This narrative review discusses the role of endoscopic ultrasound within these surveillance programs. It details its overall strengths and limitations, yield, burden on patients, and how it compares to magnetic resonance imaging. Finally, recommendations are given when and how to incorporate endoscopic ultrasound in the surveillance of high-risk individuals.
Collapse
Affiliation(s)
- Kasper A Overbeek
- Erasmus MC Cancer Institute, Department of Gastroenterology & Hepatology, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Djuna L Cahen
- Erasmus MC Cancer Institute, Department of Gastroenterology & Hepatology, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marco J Bruno
- Erasmus MC Cancer Institute, Department of Gastroenterology & Hepatology, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
31
|
Earl J, Fuentes R, Sanchez MEC, de Paredes AGG, Muñoz M, Sanjuanbenito A, Lobo E, Caminoa A, Rodríguez M, Barreto E, López JV, Ruz-Caracuel I, Durán SL, Olcina JRF, Sánchez BL, Páez SC, Torres A, Blázquez J, Sequeros EV, Carrato A. The Spanish Familial Pancreatic Cancer Registry (PANGENFAM): a decade follow-up of individuals at high-risk for pancreatic cancer. Fam Cancer 2024; 23:383-392. [PMID: 38753287 PMCID: PMC11254983 DOI: 10.1007/s10689-024-00388-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 04/04/2024] [Indexed: 07/18/2024]
Abstract
The Spanish Familial Pancreatic Cancer Registry (PANGENFAM) was established in 2009 and aims to characterize the genotype and phenotype of familial pancreatic cancer (FPC). Furthermore, an early detection screening program for pancreatic ductal adenocarcinoma (PDAC) is provided to healthy high-risk individuals from FPC and hereditary pancreatic cancer families (first-degree relatives). This article describes our experience over the last 10 years in high-risk screening. Hereditary and familial pancreatic cancer families were identified through the oncology and gastroenterology units. High-risk individuals underwent annual screening with endoscopic ultrasound (EUS) and magnetic resonance (MRI) from age 40 or 10 years younger than the youngest affected family member. Results: PANGENFAM has enrolled 290 individuals from 143 families, including 52 PDAC cases and 238 high-risk individuals. All high-risk individuals eligible for screening were offered to enter the surveillance program, with 143 currently participating. Pancreatic abnormalities were detected in 94 individuals (median age 53 years (29-83), with common findings including cystic lesions and inhomogeneous parenchyma. Imaging test concordance was 66%. Surgical intervention was performed in 4 high-risk individuals following highly suspicious lesions detected by imaging. PANGENFAM is a valuable resource for science innovation, such as biobanking, with clinical and imaging data available for analysis. For high-risk families, it may offer a potential for early diagnosis. Collaboration with other national and international registries is needed to increase our understanding of the disease biology and to standardize criteria for inclusion and follow-up, optimizing cost-effectiveness and efficacy.
Collapse
Affiliation(s)
- Julie Earl
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain.
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain.
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain.
| | - Raquel Fuentes
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
| | - María E Castillo Sanchez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
| | - Ana García García de Paredes
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - María Muñoz
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Radiology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alfonso Sanjuanbenito
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Pancreatic and Biliopancreatic Surgery Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Eduardo Lobo
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Pancreatic and Biliopancreatic Surgery Unit, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alejandra Caminoa
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Department of Pathology, Hospital Universitario Ramón y Cajal, 28034, Madrid, Spain
| | - Mercedes Rodríguez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
| | - Emma Barreto
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- University of Alcalá, Madrid, Spain
| | - Jorge Villalón López
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Ignacio Ruz-Caracuel
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- Department of Pathology, Hospital Universitario Ramón y Cajal, 28034, Madrid, Spain
| | - Sergio López Durán
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - José Ramón Foruny Olcina
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Gastroenterology and Hepatology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Bárbara Luna Sánchez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Sonia Camaño Páez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Ana Torres
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Biobank and Biomodels Platform, Spanish National Biobanks Network (ISCIII Biobank Register No. B.0000678), ISCIII Research and Development Platforms in Biomedicine and Health Sciences, BioBank Hospital Ramón y Cajal-IRYCIS, Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9, 100, PT20/004528034, Madrid, Spain
| | - Javier Blázquez
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Enrique Vázquez Sequeros
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, 28034, Madrid, Spain
| | - Alfredo Carrato
- Ramón y Cajal Health Research Institute (IRYCIS), Carretera Colmenar Km 9,100, 28034, Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- University of Alcalá, Madrid, Spain
- Pancreatic Cancer Europe, Brussels, Belgium
| |
Collapse
|
32
|
Bogdanski AM, van Hooft JE, Boekestijn B, Bonsing BA, Wasser MNJM, Klatte DCF, van Leerdam ME. Aspects and outcomes of surveillance for individuals at high-risk of pancreatic cancer. Fam Cancer 2024; 23:323-339. [PMID: 38619782 PMCID: PMC11255004 DOI: 10.1007/s10689-024-00368-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/24/2024] [Indexed: 04/16/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related deaths and is associated with a poor prognosis. The majority of these cancers are detected at a late stage, contributing to the bad prognosis. This underscores the need for novel, enhanced early detection strategies to improve the outcomes. While population-based screening is not recommended due to the relatively low incidence of PDAC, surveillance is recommended for individuals at high risk for PDAC due to their increased incidence of the disease. However, the outcomes of pancreatic cancer surveillance in high-risk individuals are not sorted out yet. In this review, we will address the identification of individuals at high risk for PDAC, discuss the objectives and targets of surveillance, outline how surveillance programs are organized, summarize the outcomes of high-risk individuals undergoing pancreatic cancer surveillance, and conclude with a future perspective on pancreatic cancer surveillance and novel developments.
Collapse
Affiliation(s)
- Aleksander M Bogdanski
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Jeanin E van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Bas Boekestijn
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin N J M Wasser
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Derk C F Klatte
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Brentnall TA. Familial pancreatic cancer: a long fruitful journey. Fam Cancer 2024; 23:217-220. [PMID: 38436765 DOI: 10.1007/s10689-024-00364-5] [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: 01/24/2024] [Accepted: 02/05/2024] [Indexed: 03/05/2024]
Abstract
In the early years of my GI fellowship, a healthy 40-year-old man came to my clinic and announced that he was going to die of pancreatic cancer. His brothers, father and uncles had all died of the disease; he felt his fate was inescapable. I asked whether his family members had seen doctors or had any tests. His answer was yes to both. Even so, doctors could not diagnose the pancreatic cancer at early stages. CT scans were always negative. I thought to myself, in order to help this patient-CT scans may not be reliable for early detection. Perhaps other methods of imaging the pancreas might be of more benefit. This patient opened a door that led to a 30-year journey of trying to detect pancreatic cancer at earlier stages when it is curable.
Collapse
Affiliation(s)
- Teresa A Brentnall
- Department of Medicine, University of Washington, PO Box 356424, 1959 NE Pacific, Seattle, WA, USA.
| |
Collapse
|
34
|
Onnekink AM, Klatte DCF, van Hooft JE, van den Berg SH, van der Zwaan SMS, van Doorn R, Hinnen SCH, Potjer TP, Bleiker EMA, van Leerdam ME. Attitudes toward genetic testing, family planning and preimplantation genetic testing in families with a germline CDKN2A pathogenic variant. Fam Cancer 2024; 23:255-265. [PMID: 38822936 PMCID: PMC11255069 DOI: 10.1007/s10689-024-00401-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/19/2024] [Indexed: 06/03/2024]
Abstract
Individuals with a germline CDKN2A pathogenic variant (PV) have a highly increased life time risk of melanoma and pancreatic cancer. This cross-sectional study assessed the attitudes among toward genetic testing, family planning, and preimplantation genetic testing (PGT) in confirmed CDKN2A PV carriers and individuals with a 50% risk of the PV (at-risk carriers) using of a one-time questionnaire.A total of 537 individuals were screened for eligibility, of whom 208 of 366 (57%) confirmed carriers (56% female, median age 54 years [IQR 46-63]) and 39 of 171 (23%) at-risk carriers (59% female, median age of 26 years [IQR 22-32]) participated in the study. Primary motivations for genetic testing were to gain control over their personal and children's cancer risk, as well as increasing cancer surveillance practices. In contrast, concerns about obtaining a mortgage and life insurance were frequently cited as reasons for postponing genetic testing. Family planning decisions remained largely unaffected in both confirmed and at-risk carriers; however, the majority of confirmed carriers were still unaware of their familial or personal cancer risk when starting a family. More than 60% of the participants were unfamiliar with PGT and only a minority (19% of confirmed carriers and 10% of at-risk carriers) would be open to considering PGT as a reproductive option. This study found different attitudes toward genetic testing, family planning, and PGT among individuals affected by the CDKN2A PV. Understanding these different attitudes can help clinicians to address the complexities surrounding these issues, especially for younger individuals facing difficult decisions about the timing of genetic testing, family planning, and the potential use of assisted reproductive options.
Collapse
Affiliation(s)
- A M Onnekink
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands.
| | - D C F Klatte
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - J E van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - S H van den Berg
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - S M S van der Zwaan
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
| | - R van Doorn
- Department of Dermatology, Leiden University Medical Center, Leiden, the Netherlands
| | - S C H Hinnen
- Department of Psycho-Oncology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Medical Psychology, Spaarne Gasthuis, Haarlem, the Netherlands
| | - T P Potjer
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - E M A Bleiker
- Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Clinical Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Gastrointestinal Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - M E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
- Department of Gastrointestinal Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
35
|
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
|
36
|
Silva-Santisteban A, Hernandez Woodbine MJ, Noriega MA, Rabinowitz LG, Grimshaw A, Farrell JJ, Chhoda A, Sawhney MS. Disparities in race, ethnicity, sex, and age inclusion in pancreatic cancer screening studies: a systematic review and meta-analysis. Gastrointest Endosc 2024; 100:1-16.e20. [PMID: 38432492 DOI: 10.1016/j.gie.2024.02.014] [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: 09/27/2023] [Revised: 02/12/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND AND AIMS Substantial differences exist in pancreatic cancer outcomes across ethnoracial stratifications. We sought to assess racial, ethnic, sex, and age reporting and inclusion of participants in pancreatic cancer screening studies. METHODS A systematic search of Cochrane Library, Ovid Embase, Google Scholar, Ovid MEDLINE, PubMed, Scopus, and Web of Science Core Collection from inception to 2022 was conducted. Original studies on pancreatic cancer screening were identified and assessed for reporting and inclusion on race, ethnicity, sex, and age. The pooled proportions of study participants for these characteristics were calculated and compared with population-based benchmarks. RESULTS Among 27 eligible pancreatic cancer screening studies, 26 reported data on either sex, race, or ethnicity, with a total of 5273 participants. Information on participant sex was reported by 26, race by 12, and ethnicity by 8 studies. Participants in these studies were almost all white (pooled proportion, 93.1%; 95% confidence interval [CI], 89.7-96.4) and non-Latino (pooled proportion, 97.4%; 95% CI, 94.0-100), and these groups were over-represented when compared with the general population. Female participants were well represented, with a pooled proportion of 63.2% (95% CI, 59.9-66.6). When reported, mean or median participant age was <60 years. Meta-regression revealed higher proportions of female participants in studies from the United States (P = .002). No association between increasing participation of racial or ethnic under-represented populations and study quality, ascending year of publication, or source of study funding was noted. CONCLUSIONS Substantial disparities in race, ethnicity, sex, and age reporting and inclusion in pancreatic cancer studies were noted, even among high-quality and publicly funded studies.
Collapse
Affiliation(s)
- Andy Silva-Santisteban
- Division of Gastroenterology & Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Maria Jose Hernandez Woodbine
- Division of Gastroenterology & Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Marco Antonio Noriega
- Division of Gastroenterology & Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Loren G Rabinowitz
- Division of Gastroenterology & Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Alyssa Grimshaw
- Cushing/Whitney Medical Library, Yale University, New Haven, Connecticut, USA
| | - James J Farrell
- Division of Gastroenterology, Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ankit Chhoda
- Division of Gastroenterology & Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Mandeep S Sawhney
- Division of Gastroenterology & Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
37
|
Qin Q, Yu R, Eriksson JE, Tsai HI, Zhu H. Cancer-associated fibroblasts in pancreatic ductal adenocarcinoma therapy: Challenges and opportunities. Cancer Lett 2024; 591:216859. [PMID: 38615928 DOI: 10.1016/j.canlet.2024.216859] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/16/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a solid organ malignancy with a high mortality rate. Statistics indicate that its incidence has been increasing as well as the associated deaths. Most patients with PDAC show poor response to therapies making the clinical management of this cancer difficult. Stromal cells in the tumor microenvironment (TME) contribute to the development of resistance to therapy in PDAC cancer cells. Cancer-associated fibroblasts (CAFs), the most prevalent stromal cells in the TME, promote a desmoplastic response, produce extracellular matrix proteins and cytokines, and directly influence the biological behavior of cancer cells. These multifaceted effects make it difficult to eradicate tumor cells from the body. As a result, CAF-targeting synergistic therapeutic strategies have gained increasing attention in recent years. However, due to the substantial heterogeneity in CAF origin, definition, and function, as well as high plasticity, majority of the available CAF-targeting therapeutic approaches are not effective, and in some cases, they exacerbate disease progression. This review primarily elucidates on the effect of CAFs on therapeutic efficiency of various treatment modalities, including chemotherapy, radiotherapy, immunotherapy, and targeted therapy. Strategies for CAF targeting therapies are also discussed.
Collapse
Affiliation(s)
- Qin Qin
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China
| | - Rong Yu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China
| | - John E Eriksson
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, FI-20520 Finland
| | - Hsiang-I Tsai
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| |
Collapse
|
38
|
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. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595399. [PMID: 38826212 PMCID: PMC11142185 DOI: 10.1101/2024.05.22.595399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
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
|
39
|
Di Y, Song J, Wang Y, Meng L. Survival impact of radiotherapy in unresectable locally advanced pancreatic cancer. Asian J Surg 2024:S1015-9584(24)00651-1. [PMID: 38599965 DOI: 10.1016/j.asjsur.2024.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/03/2024] [Indexed: 04/12/2024] Open
Affiliation(s)
- Yupeng Di
- Department of Radiation Oncology, Air Force Medical Center, PLA, Beijing, China
| | - Jiazhao Song
- Department of Radiation Oncology, Air Force Medical Center, PLA, Beijing, China
| | - Yingjie Wang
- Department of Radiation Oncology, Air Force Medical Center, PLA, Beijing, China.
| | - Lingling Meng
- Department of Radiation Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China.
| |
Collapse
|
40
|
Ben-Ami R, Wang QL, Zhang J, Supplee JG, Fahrmann JF, Lehmann-Werman R, Brais LK, Nowak J, Yuan C, Loftus M, Babic A, Irajizad E, Davidi T, Zick A, Hubert A, Neiman D, Piyanzin S, Gal-Rosenberg O, Horn A, Shemer R, Glaser B, Boos N, Jajoo K, Lee L, Clancy TE, Rubinson DA, Ng K, Chabot JA, Kastrinos F, Kluger M, Aguirre AJ, Jänne PA, Bardeesy N, Stanger B, O'Hara MH, Till J, Maitra A, Carpenter EL, Bullock AJ, Genkinger J, Hanash SM, Paweletz CP, Dor Y, Wolpin BM. Protein biomarkers and alternatively methylated cell-free DNA detect early stage pancreatic cancer. Gut 2024; 73:639-648. [PMID: 38123998 PMCID: PMC10958271 DOI: 10.1136/gutjnl-2023-331074] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/26/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma (PDAC) is commonly diagnosed at an advanced stage. Liquid biopsy approaches may facilitate detection of early stage PDAC when curative treatments can be employed. DESIGN To assess circulating marker discrimination in training, testing and validation patient cohorts (total n=426 patients), plasma markers were measured among PDAC cases and patients with chronic pancreatitis, colorectal cancer (CRC), and healthy controls. Using CA19-9 as an anchor marker, measurements were made of two protein markers (TIMP1, LRG1) and cell-free DNA (cfDNA) pancreas-specific methylation at 9 loci encompassing 61 CpG sites. RESULTS Comparative methylome analysis identified nine loci that were differentially methylated in exocrine pancreas DNA. In the training set (n=124 patients), cfDNA methylation markers distinguished PDAC from healthy and CRC controls. In the testing set of 86 early stage PDAC and 86 matched healthy controls, CA19-9 had an area under the receiver operating characteristic curve (AUC) of 0.88 (95% CI 0.83 to 0.94), which was increased by adding TIMP1 (AUC 0.92; 95% CI 0.88 to 0.96; p=0.06), LRG1 (AUC 0.92; 95% CI 0.88 to 0.96; p=0.02) or exocrine pancreas-specific cfDNA methylation markers at nine loci (AUC 0.92; 95% CI 0.88 to 0.96; p=0.02). In the validation set of 40 early stage PDAC and 40 matched healthy controls, a combined panel including CA19-9, TIMP1 and a 9-loci cfDNA methylation panel had greater discrimination (AUC 0.86, 95% CI 0.77 to 0.95) than CA19-9 alone (AUC 0.82; 95% CI 0.72 to 0.92). CONCLUSION A combined panel of circulating markers including proteins and methylated cfDNA increased discrimination compared with CA19-9 alone for early stage PDAC.
Collapse
Affiliation(s)
- Roni Ben-Ami
- Department of Developmental Biology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Qiao-Li Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Jinming Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Julianna G Supplee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Johannes F Fahrmann
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Roni Lehmann-Werman
- Department of Developmental Biology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lauren K Brais
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan Nowak
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Maureen Loftus
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Ehsan Irajizad
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tal Davidi
- Sharett Institute of Oncology, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Aviad Zick
- Sharett Institute of Oncology, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Ayala Hubert
- Sharett Institute of Oncology, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Daniel Neiman
- Department of Developmental Biology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sheina Piyanzin
- Department of Developmental Biology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ofer Gal-Rosenberg
- Department of Developmental Biology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Amit Horn
- Department of Developmental Biology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ruth Shemer
- Department of Developmental Biology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Benjamin Glaser
- Department of Developmental Biology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Endocrinology and Metabolism, Hadassah Medical Center, Jerusalem, Israel
| | - Natalia Boos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Kunal Jajoo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Linda Lee
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas E Clancy
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Douglas A Rubinson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - John A Chabot
- Department of Surgery, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, USA
| | - Fay Kastrinos
- Division of Digestive and Liver Diseases, Columbia University Irving Medical Cancer and the Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Michael Kluger
- Department of Surgery, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, USA
| | - Andrew J Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Pasi A Jänne
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center, Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ben Stanger
- Department of Medicine, Division of Gastroenterology, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Mark H O'Hara
- Department of Medicine, Division of Hematology-Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jacob Till
- Department of Medicine, Division of Hematology-Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anirban Maitra
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Erica L Carpenter
- Department of Medicine, Division of Hematology-Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andrea J Bullock
- Division of Hematology and Oncology, Beth-Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Jeanine Genkinger
- Department of epidemiology, Mailman school of public health, Columbia university, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia university Irving Medical Center, New York, New York, USA
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Cloud P Paweletz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
41
|
Gao L, Ugalde A, Livingston PM, White V, Watts JJ, Jongebloed H, McCaffrey N, Menzies D, Robinson S. Simulating the healthcare workforce impact and capacity for pancreatic cancer care in Victoria: a model-based analysis. BMC Health Serv Res 2024; 24:239. [PMID: 38395852 PMCID: PMC10893744 DOI: 10.1186/s12913-024-10722-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND The incidence of pancreatic cancer is rising. With improvements in knowledge for screening and early detection, earlier detection of pancreatic cancer will continue to be more common. To support workforce planning, our aim is to perform a model-based analysis that simulates the potential impact on the healthcare workforce, assuming an earlier diagnosis of pancreatic cancer. METHODS We developed a simulation model to estimate the demand (i.e. new cases of pancreatic cancer) and supply (i.e. the healthcare workforce including general surgeons, medical oncologists, radiation oncologists, pain medicine physicians, and palliative care physicians) between 2023 and 2027 in Victoria, Australia. The model compares the current scenario to one in which pancreatic cancer is diagnosed at an earlier stage. The incidence of pancreatic cancer in Victoria, five-year survival rates, and Victoria's population size were obtained from Victorian Cancer Registry, Cancer Council NSW, and Australian Bureau of Statistics respectively. The healthcare workforce data were sourced from the Australian Government Department of Health and Aged Care's Health Workforce Data. The model was constructed at the remoteness level. We analysed the new cases and the number of healthcare workforce by profession together to assess the impact on the healthcare workforce. RESULTS In the status quo, over the next five years, there will be 198 to 220 stages I-II, 297 to 330 stage III, and 495 to 550 stage IV pancreatic cancer cases diagnosed annually, respectively. Assuming 20-70% of the shift towards pancreatic cancer's earlier diagnosis (shifting from stage IV to stages I-II pancreatic cancer within one year), the stages I-II cases could increase to 351 to 390 or 598 to 665 per year. The shift to early diagnosis led to substantial survival gains, translating into an additional 284 or 795 out of 5246 patients with pancreatic cancer remaining alive up to year 5 post-diagnosis. Workforce supply decreases significantly by the remoteness levels, and remote areas face a shortage of key medical professionals registered in delivering pancreatic cancer care, suggesting travel necessities by patients or clinicians. CONCLUSION Improving the early detection and diagnosis of pancreatic cancer is expected to bring significant survival benefits, although there are workforce distribution imbalances in Victoria that may affect the ability to achieve the anticipated survival gain.
Collapse
Affiliation(s)
- Lan Gao
- Deakin Health Economics, Institute of Health Transformation, Faculty of Health, Deakin University, 1 Gheringhap St, 3220, Geelong, Australia.
| | - Anna Ugalde
- School of Nursing & Midwifery, Institute of Health Transformation, Faculty of Health, Deakin University, Melbourne, Australia
| | - Patricia M Livingston
- School of Nursing & Midwifery, Institute of Health Transformation, Faculty of Health, Deakin University, Melbourne, Australia
| | - Victoria White
- School of Nursing & Midwifery, Institute of Health Transformation, Faculty of Health, Deakin University, Melbourne, Australia
| | - Jennifer J Watts
- Deakin Health Economics, Institute of Health Transformation, Faculty of Health, Deakin University, 1 Gheringhap St, 3220, Geelong, Australia
| | - Hannah Jongebloed
- School of Nursing & Midwifery, Institute of Health Transformation, Faculty of Health, Deakin University, Melbourne, Australia
| | - Nikki McCaffrey
- Deakin Health Economics, Institute of Health Transformation, Faculty of Health, Deakin University, 1 Gheringhap St, 3220, Geelong, Australia
| | | | - Suzanne Robinson
- Deakin Health Economics, Institute of Health Transformation, Faculty of Health, Deakin University, 1 Gheringhap St, 3220, Geelong, Australia
| |
Collapse
|
42
|
Peters MLB, Eckel A, Seguin CL, Davidi B, Howard DH, Knudsen AB, Pandharipande PV. Cost-Effectiveness Analysis of Screening for Pancreatic Cancer Among High-Risk Populations. JCO Oncol Pract 2024; 20:278-290. [PMID: 38086003 PMCID: PMC10911581 DOI: 10.1200/op.23.00495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/05/2023] [Accepted: 10/30/2023] [Indexed: 12/19/2023] Open
Abstract
PURPOSE We evaluated the potential cost-effectiveness of combined magnetic resonance imaging (MRI) and endoscopic ultrasound (EUS) screening for pancreatic ductal adenocarcinoma (PDAC) among populations at high risk for the disease. METHODS We used a microsimulation model of the natural history of PDAC to estimate the lifetime health benefits, costs, and cost-effectiveness of PDAC screening among populations with specific genetic risk factors for PDAC, including BRCA1 and BRCA2, PALB2, ATM, Lynch syndrome, TP53, CDKN2A, and STK11. For each high-risk population, we simulated 29 screening strategies, defined by starting age and frequency. Screening included MRI with follow-up EUS in a subset of patients. Costs of tests were based on Medicare reimbursement for MRI, EUS, fine-needle aspiration biopsy, and pancreatectomy. Cancer-related cost by stage of disease and phase of treatment was based on the literature. For each high-risk population, we performed an incremental cost-effectiveness analysis, assuming a willingness-to-pay (WTP) threshold of $100,000 US dollars (USD) per quality-adjusted life year (QALY) gained. RESULTS For men with relative risk (RR) 12.33 (CDKN2A) and RR 28 (STK11), annual screening was cost-effective, starting at age 55 and 40 years, respectively. For women, screening was only cost-effective for those with RR 28 (STK11), with annual screening starting at age 45 years. CONCLUSION Combined MRI/EUS screening may be a cost-effective approach for the highest-risk populations (among mutations considered, those with RR >12). However, for those with moderate risk (RR, 5-12), screening would only be cost-effective at higher WTP thresholds (eg, $200K USD/QALY) or with once-only screening.
Collapse
Affiliation(s)
- Mary Linton B. Peters
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA
| | - Andrew Eckel
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA
| | - Claudia L. Seguin
- Department of Radiology, The Ohio State University College of Medicine, Columbus, OH
| | - Barak Davidi
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA
| | - David H. Howard
- Department of Health Policy and Management, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Amy B. Knudsen
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, MA
- Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Pari V. Pandharipande
- Department of Radiology, The Ohio State University College of Medicine, Columbus, OH
| |
Collapse
|
43
|
Laish I, Schechter M, Dancour A, Lieberman S, Levi Z, Goldberg Y, Kedar I, Hasnis E, Half E, Levi GR, Katz L, Vainer ED, Genzel D, Aharoni M, Chen-Shtoyerman R, Abu-Freha N, Raitses-Gurevich M, Golan T, Bernstein-Molho R, Ben Yehoyada M, Gluck N, Rosner G. The benefit of pancreatic cancer surveillance in carriers of germline BRCA1/2 pathogenic variants. Cancer 2024; 130:256-266. [PMID: 37861363 DOI: 10.1002/cncr.35052] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/22/2023] [Accepted: 08/03/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Surveillance of high-risk individuals for pancreatic ductal adenocarcinoma (PDAC) is recommended. This study aimed to determine the prevalence and outcomes of PDAC and its precursor lesions in BRCA1/2 pathogenic variants (PVs) carriers undergoing pancreatic surveillance. METHODS A retrospective multicenter cohort study of pancreatic surveillance outcomes in Israeli BRCA1/2 carriers preferably with a family history of PDAC. RESULTS A total of 180 asymptomatic carriers participated in the screening programs, including 57 (31.7%) with BRCA1 PVs, 121 (67.2%) with BRCA2 PVs, and 12 (6.6%) with PVs in BRCA1/2 and other genes, for a median follow-up period of 4 years. Ninety-one individuals (50.5%) fulfilled the International Cancer of the Pancreas Screening (CAPS) criteria for surveillance whereas 116 (64.4%) fulfilled the American College of Gastroenterology (ACG) criteria. There were four cases of adenocarcinoma and four cases of grade 1-neuroendocrine tumor (G1-NET). All were BRCA2 carriers, and two had no family history of PDAC. Three cancer patients were at resectable stages (IA, IIA, IIB) whereas one had a stage IIIB tumor. Of the G1-NET cases, one had surgery and the others were only followed. Success rate for detection of confined pancreatic carcinoma was thus 1.6% (three of 180) in the whole cohort, 1.6% (two of 116) among individuals who fulfilled ACG criteria and 2.2% (two of 91) in those fulfilling CAPS criteria for surveillance. CONCLUSIONS Despite the low detection rate of PDAC and its' high-risk neoplastic precursor lesions among BRCA1/2 carriers undergoing pancreatic surveillance, 75% of cancer cases were detected at a resectable stage.
Collapse
Affiliation(s)
- Ido Laish
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Menachem Schechter
- Gastroenterology Institute, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Alain Dancour
- Gastroenterology Institute, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Sari Lieberman
- Medical Genetics Institute, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Zohar Levi
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Gastroenterology Institute, Beilinson Hospital, Petah Tikva, Israel
| | - Yael Goldberg
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Raphael Recanati Genetics Institute, Beilinson Hospital, Petah Tikva, Israel
| | - Inbal Kedar
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Raphael Recanati Genetics Institute, Beilinson Hospital, Petah Tikva, Israel
| | - Erez Hasnis
- Gastroenterology Institute, Rambam Health Care Campus, Haifa, Israel
| | - Elizabeth Half
- Gastroenterology Institute, Rambam Health Care Campus, Haifa, Israel
| | | | - Lior Katz
- Section of Gastroenterology and Hepatology, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Elez D Vainer
- Section of Gastroenterology and Hepatology, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Dor Genzel
- Section of Gastroenterology and Hepatology, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Maya Aharoni
- Gastroenterology Institute, Kaplan Medical Center, Rehovot, Israel
| | - Rakefet Chen-Shtoyerman
- The Genetic Institute, Kaplan Medical Center, Rehovot, Israel
- The Adelson School of Medicine and the Molecular Biology Department, Ariel University, Ariel, Israel
| | - Naim Abu-Freha
- The Institute of Gastroenterology and Hepatology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Maria Raitses-Gurevich
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Talia Golan
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Rinat Bernstein-Molho
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Danek Gertner Institute of Human Genetics, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Merav Ben Yehoyada
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Nathan Gluck
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Guy Rosner
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| |
Collapse
|
44
|
Paiella S, Secchettin E, Lionetto G, Archibugi L, Azzolina D, Casciani F, Simeone DM, Overbeek KA, Goggins M, Farrell J, Ponz de Leon Pisani R, Tridenti M, Corciulo MA, Malleo G, Arcidiacono PG, Falconi M, Gregori D, Bassi C, Salvia R, Capurso G. Surveillance of Individuals at High Risk of Developing Pancreatic Cancer: A Prevalence Meta-analysis to Estimate the Rate of Low-yield Surgery. Ann Surg 2024; 279:37-44. [PMID: 37681303 DOI: 10.1097/sla.0000000000006094] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
OBJECTIVE To quantify the rate of low-yield surgery, defined as no high-grade dysplastic precursor lesions or T1N0M0 pancreatic cancer at pathology, during pancreatic cancer surveillance. BACKGROUND Global efforts have been made in pancreatic cancer surveillance to anticipate the diagnosis of pancreatic cancer at an early stage and improve survival in high-risk individuals (HRIs) with a hereditary predisposition. The negative impact of pancreatic cancer surveillance when surgery is performed for low-grade dysplasia or a non-neoplastic condition is not well quantified. MATERIALS AND METHODS A systematic search and prevalence meta-analysis was performed for studies reporting surgery with final diagnoses other than those defined by the Cancer of the Pancreas Screening (CAPS) goals from January 2000 to July 2023. The secondary outcome was the pooled proportion of final diagnoses matching the CAPS goals (PROSPERO: #CRD42022300408). RESULTS Twenty-three articles with 5027 patients (median 109 patients/study, interquartile range 251) were included. The pooled prevalence of low-yield surgery was 2.1% (95% CI: 0.9-3.7, I2 : 83%). In the subgroup analysis, this prevalence was nonsignificantly higher in studies that only included familial pancreatic cancer subjects without known pathogenic variants, compared with those enrolling pathogenic variant carriers. No effect modifiers were found. Overall, the pooled prevalence of subjects under surveillance who had a pancreatic resection that contained target lesions was 0.8% (95% CI, 0.3-1.5, I2 : 24%]. The temporal analysis showed that the rate of low-yield surgeries decreased in the last decades and stabilized at around 1% (test for subgroup differences P <0.01). CONCLUSIONS The risk of "low-yield" surgery during pancreatic cancer surveillance is relatively low but should be thoroughly discussed with individuals under surveillance.
Collapse
Affiliation(s)
- Salvatore Paiella
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Erica Secchettin
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Gabriella Lionetto
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Livia Archibugi
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| | - Danila Azzolina
- Department of Environmental and Preventive Science, University of Ferrara, Ferrara, Italy
| | - Fabio Casciani
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Diane M Simeone
- Department of Surgery, New York University, New York, NY
- Perlmutter Cancer Center, New York University, New York, NY
| | - Kasper A Overbeek
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Michael Goggins
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - James Farrell
- Yale Center for Pancreatic Disease, Yale University School of Medicine, New Haven, CT
| | - Ruggero Ponz de Leon Pisani
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| | - Maddalena Tridenti
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| | - Maria Assunta Corciulo
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, Padova, Italy
| | - Giuseppe Malleo
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Paolo Giorgio Arcidiacono
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| | - Massimo Falconi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Pancreatic Surgery and Transplantation Unit, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Dario Gregori
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, Padova, Italy
| | - Claudio Bassi
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Roberto Salvia
- General and Pancreatic Surgery Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan, Italy
| |
Collapse
|
45
|
Wu BU, Chen Q, Moon BH, Lustigova E, Nielsen EG, Alvarado M, Ahmed SA. Association of Glycated Hemoglobin With a Risk of Pancreatic Cancer in High-Risk Individuals Based on Genetic and Family History. Clin Transl Gastroenterol 2024; 15:e00650. [PMID: 37800692 PMCID: PMC10810597 DOI: 10.14309/ctg.0000000000000650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/21/2023] [Indexed: 10/07/2023] Open
Abstract
INTRODUCTION Screening for pancreatic cancer (PC) is suggested for high-risk individuals. Additional risk factors may enhance early detection in this population. METHODS Retrospective cohort study among patients with germline variants and/or familial pancreatic cancer in an integrated healthcare system between 2003 and 2019. We calculated the incidence rate (IR) by risk category and performed a nested case-control study to evaluate the relationship between HbA1C and PC within 3 years before diagnosis (cases) or match date (controls). Cases were matched 1:4 by age, sex, and timing of HbA1c. Logistic regression was performed to assess an independent association with PC. RESULTS We identified 5,931 high-risk individuals: 1,175(19.8%) familial PC, 45(0.8%) high-risk germline variants ( STK11, CDKN2A ), 4,097(69.1%) had other germline variants ( ATM, BRCA 1, BRCA 2, CASR, CDKN2A, CFTR, EPCAM, MLH1, MSH2, MSH6, PALB2, PRSS1, STK11, and TP53 ), and 614(10.4%) had both germline variants and family history. Sixty-eight patients (1.1%) developed PC; 50% were metastatic at diagnosis. High-risk variant was associated with greatest risk of PC, IR = 85.1(95% confidence interval: 36.7-197.6)/10,000 person-years; other germline variants and first-degree relative had IR = 33 (18.4, 59.3), whereas IR among ≥2 first-degree relative alone was 10.7 (6.1, 18.8). HbA1c was significantly higher among cases vs controls (median = 7.0% vs 6.4%, P = 0.02). In multivariable analysis, every 1% increase in HbA1c was associated with 36% increase in odds of PC (odds ratio 1.36, 95% confidence interval: 1.08-1.72). Pancreatitis was independently associated with a risk of PC (odds ratio 3.93, 95% confidence limit 1.19, 12.91). DISCUSSION Risk of PC varies among high-risk individuals. HbA1c and history of pancreatitis may be useful additional markers for early detection in this patient population.
Collapse
Affiliation(s)
- Bechien U. Wu
- Center for Pancreatic Care, Division of Gastroenterology, Kaiser Permanente Los Angeles Medical Center, Los Angeles, California, USA;
| | - Qiaoling Chen
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California, USA;
| | - Becky H. Moon
- Center for Pancreatic Care, Division of Gastroenterology, Kaiser Permanente Los Angeles Medical Center, Los Angeles, California, USA;
| | - Eva Lustigova
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, California, USA;
| | - Erin G. Nielsen
- Department of Genetics, Southern California Medical Group, Pasadena, California, USA;
| | - Monica Alvarado
- Department of Genetics, Southern California Medical Group, Pasadena, California, USA;
| | - Syed A. Ahmed
- Department of Genetics, Kaiser Permanente Riverside Medical Center, Riverside, California, USA
| |
Collapse
|
46
|
Anez-Bruzual I, Coughlin S, Clay D, Heiman J, Dungan M, Weber M, Almario CV, Leung G, Ahmad NA, Ginsberg GG, Kochman ML, Valverde KD, Long JM, Katona BW. Assessment of the Psychosocial Impact of Pancreatic Cancer Surveillance in High-Risk Individuals. Cancers (Basel) 2023; 16:86. [PMID: 38201514 PMCID: PMC10777978 DOI: 10.3390/cancers16010086] [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: 10/15/2023] [Revised: 12/05/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
OBJECTIVES Pancreatic cancer (PC) surveillance of high-risk individuals (HRIs) downstages PC and improves survival. However, it remains less clear whether PC surveillance has a positive psychosocial impact on HRIs. Herein, we aimed to define the attitudes and beliefs of HRIs undergoing PC surveillance, and the immediate and sustained psychosocial impact of PC surveillance in HRIs. METHODS 100 HRIs undergoing PC surveillance by endoscopic ultrasound (EUS) completed three surveys addressing different components of the psychosocial impact of PC surveillance. Logistic regression analyses were performed to identify predictive factors relating to these components. RESULTS Most HRIs reported increased perceived benefits of PC surveillance, self-efficacy, and perceived severity of PC. HRIs reported few negative emotions prior to surveillance and frequent positive emotions after surveillance. Compared to prior to surveillance, there was a 53.5% decrease in the level of distress reported by HRIs after surveillance, which was sustained for 4-6 weeks post-surveillance. Family history of PC and lower self-reported mental health were identified as predictors for increased perceived susceptibility to PC (p < 0.01) and greater change in distress pre- to post-surveillance (p < 0.01), respectively. CONCLUSIONS Our findings suggest that PC surveillance can lead to sustained psychosocial benefits in HRIs.
Collapse
Affiliation(s)
- Isabel Anez-Bruzual
- Master of Science in Genetic Counseling Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (I.A.-B.); (K.D.V.)
| | - Sarah Coughlin
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.C.); (D.C.); (J.H.); (M.D.); (M.W.); (G.L.); (N.A.A.); (G.G.G.); (M.L.K.)
| | - Daniel Clay
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.C.); (D.C.); (J.H.); (M.D.); (M.W.); (G.L.); (N.A.A.); (G.G.G.); (M.L.K.)
| | - Jordan Heiman
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.C.); (D.C.); (J.H.); (M.D.); (M.W.); (G.L.); (N.A.A.); (G.G.G.); (M.L.K.)
| | - Michaela Dungan
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.C.); (D.C.); (J.H.); (M.D.); (M.W.); (G.L.); (N.A.A.); (G.G.G.); (M.L.K.)
| | - Marina Weber
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.C.); (D.C.); (J.H.); (M.D.); (M.W.); (G.L.); (N.A.A.); (G.G.G.); (M.L.K.)
| | - Christopher V. Almario
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Galen Leung
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.C.); (D.C.); (J.H.); (M.D.); (M.W.); (G.L.); (N.A.A.); (G.G.G.); (M.L.K.)
| | - Nuzhat A. Ahmad
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.C.); (D.C.); (J.H.); (M.D.); (M.W.); (G.L.); (N.A.A.); (G.G.G.); (M.L.K.)
| | - Gregory G. Ginsberg
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.C.); (D.C.); (J.H.); (M.D.); (M.W.); (G.L.); (N.A.A.); (G.G.G.); (M.L.K.)
| | - Michael L. Kochman
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.C.); (D.C.); (J.H.); (M.D.); (M.W.); (G.L.); (N.A.A.); (G.G.G.); (M.L.K.)
| | - Kathleen D. Valverde
- Master of Science in Genetic Counseling Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (I.A.-B.); (K.D.V.)
| | - Jessica M. Long
- Division of Hematology-Oncology, Department of Medicine, Penn Medicine, Philadelphia, PA 19104, USA;
| | - Bryson W. Katona
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.C.); (D.C.); (J.H.); (M.D.); (M.W.); (G.L.); (N.A.A.); (G.G.G.); (M.L.K.)
| |
Collapse
|
47
|
Sirtl S, Vornhülz M, Hofmann FO, Mayerle J, Beyer G. [Pancreatic cancer-screening or surveillance?]. RADIOLOGIE (HEIDELBERG, GERMANY) 2023; 63:908-915. [PMID: 37878016 DOI: 10.1007/s00117-023-01227-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
BACKGROUND Despite continuous improvement of diagnostic and therapeutic procedures, the number of new pancreatic ductal adenocarcinoma (PDAC) cases diagnosed annually almost equals the number of PDAC-related deaths. Prerequisite for curative treatment is a resectable tumor at the time of diagnosis. Individuals with genetic and/or familial risk profiles should therefore be screened and included in structured surveillance programs. OBJECTIVES Description of the status quo and usefulness of current PDAC screening and surveillance concepts. METHODS A selective literature search of current national and international guidelines including underlying literature was performed. RESULTS Nearly half of pancreatic cancer cases are missed by currently available surveillance programs, even in high-risk cohorts. Magnetic resonance imaging and endoscopic ultrasound supplemented by CA19‑9 (± HbA1c) are not accurate enough to ensure robust earlier pancreatic cancer detection. Complementary biomarker panels will take on a crucial diagnostic role in the future.
Collapse
Affiliation(s)
- Simon Sirtl
- Medizinische Klinik und Poliklinik II, LMU Klinikum, 81377, München, Deutschland.
| | - Marlies Vornhülz
- Medizinische Klinik und Poliklinik II, LMU Klinikum, 81377, München, Deutschland
| | - Felix O Hofmann
- Klinik für Allgemein‑, Viszeral- und Transplantationschirurgie, LMU Klinikum, München, Deutschland
| | - Julia Mayerle
- Medizinische Klinik und Poliklinik II, LMU Klinikum, 81377, München, Deutschland.
| | - Georg Beyer
- Medizinische Klinik und Poliklinik II, LMU Klinikum, 81377, München, Deutschland
| |
Collapse
|
48
|
Koopmann BDM, Dunnewind N, van Duuren LA, Lansdorp-Vogelaar I, Naber SK, Cahen DL, Bruno MJ, de Kok IMCM. The Natural Disease Course of Pancreatic Cyst-Associated Neoplasia, Dysplasia, and Ductal Adenocarcinoma: Results of a Microsimulation Model. Gastroenterology 2023; 165:1522-1532. [PMID: 37633497 DOI: 10.1053/j.gastro.2023.08.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/19/2023] [Accepted: 08/11/2023] [Indexed: 08/28/2023]
Abstract
BACKGROUND & AIMS Estimates on the progression of precursor lesions to pancreatic cancer (PC) are scarce. We used microsimulation modeling to gain insight into the natural disease course of PC and its precursors. This information is pivotal to explore the efficacy of PC screening. METHODS A Microsimulation Screening Analysis model was developed in which pancreatic intraepithelial neoplasms and cysts can evolve from low-grade dysplasia (LGD) to high-grade dysplasia (HGD) to PC. The model was calibrated to Dutch PC incidence data and Japanese precursor prevalence data (autopsy cases without PC) and provides estimates of PC progression (precursor lesion onset and stage duration). RESULTS Mean LGD state durations of cysts and pancreatic intraepithelial neoplasms were 15.8 years and 17.1 years, respectively. Mean HGD state duration was 5.8 years. For lesions that progress to PC, the mean duration was 4.8-4.9 years for LGD lesions and 4.0-4.1 years for HGD lesions. In 13.7% of individuals who developed PC, the HGD state lasted less than 1 year. The probability that an individual at age 50 years developed PC in the next 20 years was estimated to be 1.8% in the presence of any cyst and 6.1% in case of an LGD mucinous cyst. This 20-year PC risk was estimated to be 5.1% for individuals with an LGD pancreatic intraepithelial neoplasm. CONCLUSIONS Mean duration of HGD lesions before development of PC was estimated to be 4.0 years. This implies a window of opportunity for screening, presuming the availability of a reliable diagnostic test. The probability that an LGD cyst will progress to cancer was predicted to be low.
Collapse
Affiliation(s)
- Brechtje D M Koopmann
- Department of Public Health, Erasmus Medical Center, Rotterdam, The Netherlands; Department of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Niels Dunnewind
- Department of Public Health, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Luuk A van Duuren
- Department of Public Health, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Steffie K Naber
- Department of Public Health, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Djuna L Cahen
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marco J Bruno
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Inge M C M de Kok
- Department of Public Health, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
49
|
Cao K, Xia Y, Yao J, Han X, Lambert L, Zhang T, Tang W, Jin G, Jiang H, Fang X, Nogues I, Li X, Guo W, Wang Y, Fang W, Qiu M, Hou Y, Kovarnik T, Vocka M, Lu Y, Chen Y, Chen X, Liu Z, Zhou J, Xie C, Zhang R, Lu H, Hager GD, Yuille AL, Lu L, Shao C, Shi Y, Zhang Q, Liang T, Zhang L, Lu J. Large-scale pancreatic cancer detection via non-contrast CT and deep learning. Nat Med 2023; 29:3033-3043. [PMID: 37985692 PMCID: PMC10719100 DOI: 10.1038/s41591-023-02640-w] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 10/12/2023] [Indexed: 11/22/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most deadly solid malignancy, is typically detected late and at an inoperable stage. Early or incidental detection is associated with prolonged survival, but screening asymptomatic individuals for PDAC using a single test remains unfeasible due to the low prevalence and potential harms of false positives. Non-contrast computed tomography (CT), routinely performed for clinical indications, offers the potential for large-scale screening, however, identification of PDAC using non-contrast CT has long been considered impossible. Here, we develop a deep learning approach, pancreatic cancer detection with artificial intelligence (PANDA), that can detect and classify pancreatic lesions with high accuracy via non-contrast CT. PANDA is trained on a dataset of 3,208 patients from a single center. PANDA achieves an area under the receiver operating characteristic curve (AUC) of 0.986-0.996 for lesion detection in a multicenter validation involving 6,239 patients across 10 centers, outperforms the mean radiologist performance by 34.1% in sensitivity and 6.3% in specificity for PDAC identification, and achieves a sensitivity of 92.9% and specificity of 99.9% for lesion detection in a real-world multi-scenario validation consisting of 20,530 consecutive patients. Notably, PANDA utilized with non-contrast CT shows non-inferiority to radiology reports (using contrast-enhanced CT) in the differentiation of common pancreatic lesion subtypes. PANDA could potentially serve as a new tool for large-scale pancreatic cancer screening.
Collapse
Affiliation(s)
- Kai Cao
- Department of Radiology, Shanghai Institution of Pancreatic Disease, Shanghai, China
| | - Yingda Xia
- DAMO Academy, Alibaba Group, New York, NY, USA
| | - Jiawen Yao
- Hupan Laboratory, Hangzhou, China
- Damo Academy, Alibaba Group, Hangzhou, China
| | - Xu Han
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Lukas Lambert
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Tingting Zhang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Tang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Gang Jin
- Department of Surgery, Shanghai Institution of Pancreatic Disease, Shanghai, China
| | - Hui Jiang
- Department of Pathology, Shanghai Institution of Pancreatic Disease, Shanghai, China
| | - Xu Fang
- Department of Radiology, Shanghai Institution of Pancreatic Disease, Shanghai, China
| | - Isabella Nogues
- Department of Biostatistics, Harvard University T.H. Chan School of Public Health, Cambridge, MA, USA
| | - Xuezhou Li
- Department of Radiology, Shanghai Institution of Pancreatic Disease, Shanghai, China
| | - Wenchao Guo
- Hupan Laboratory, Hangzhou, China
- Damo Academy, Alibaba Group, Hangzhou, China
| | - Yu Wang
- Hupan Laboratory, Hangzhou, China
- Damo Academy, Alibaba Group, Hangzhou, China
| | - Wei Fang
- Hupan Laboratory, Hangzhou, China
- Damo Academy, Alibaba Group, Hangzhou, China
| | - Mingyan Qiu
- Hupan Laboratory, Hangzhou, China
- Damo Academy, Alibaba Group, Hangzhou, China
| | - Yang Hou
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tomas Kovarnik
- Department of Invasive Cardiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Michal Vocka
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Yimei Lu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yingli Chen
- Department of Surgery, Shanghai Institution of Pancreatic Disease, Shanghai, China
| | - Xin Chen
- Department of Radiology, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Jian Zhou
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Chuanmiao Xie
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Rong Zhang
- Department of Radiology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Hong Lu
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Gregory D Hager
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Alan L Yuille
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Le Lu
- DAMO Academy, Alibaba Group, New York, NY, USA
| | - Chengwei Shao
- Department of Radiology, Shanghai Institution of Pancreatic Disease, Shanghai, China.
| | - Yu Shi
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Zhejiang University, Hangzhou, China.
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Zhejiang University, Hangzhou, China.
| | - Ling Zhang
- DAMO Academy, Alibaba Group, New York, NY, USA.
| | - Jianping Lu
- Department of Radiology, Shanghai Institution of Pancreatic Disease, Shanghai, China.
| |
Collapse
|
50
|
Mertowska P, Mertowski S, Smolak K, Pasiarski M, Smok-Kalwat J, Góźdź S, Grywalska E. Exploring the Significance of Immune Checkpoints and EBV Reactivation in Antibody Deficiencies with Near-Normal Immunoglobulin Levels or Hyperimmunoglobulinemia. Cancers (Basel) 2023; 15:5059. [PMID: 37894426 PMCID: PMC10605741 DOI: 10.3390/cancers15205059] [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: 08/15/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
This study delves into the intricate landscape of primary immunodeficiencies, with a particular focus on antibody deficiencies characterized by near-normal immunoglobulin levels or hyperimmunoglobulinemia. Contrary to the conventional focus on genetic dysregulation, these studies investigate the key roles of immune checkpoints, such as PD-1/PD-L1, CTLA-4/CD86, and CD200R/CD200, on selected subpopulations of T and B lymphocytes and their serum concentrations of soluble forms in patients recruited for the studies in healthy volunteers. In addition, the studies also show the role of Epstein-Barr virus (EBV) reactivation and interactions with tested pathways of immune checkpoints involved in the immunopathogenesis of this disease. By examining the context of antibody deficiencies, this study sheds light on the nuanced interplay of factors beyond genetics, particularly the immune dysregulations that occur in the course of this type of disease and the potential role of EBV reactivation, which affects the clinical presentation of patients and may contribute to the development of cancer in the future, especially related to hematological malignancies.
Collapse
Affiliation(s)
- Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (P.M.); (E.G.)
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (P.M.); (E.G.)
| | - Konrad Smolak
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (P.M.); (E.G.)
| | - Marcin Pasiarski
- Department of Immunology, Faculty of Health Sciences, Jan Kochanowski University, 25-317 Kielce, Poland;
- Department of Hematology, Holy Cross Cancer Centre, 25-734 Kielce, Poland; (J.S.-K.); (S.G.)
| | - Jolanta Smok-Kalwat
- Department of Hematology, Holy Cross Cancer Centre, 25-734 Kielce, Poland; (J.S.-K.); (S.G.)
| | - Stanisław Góźdź
- Department of Hematology, Holy Cross Cancer Centre, 25-734 Kielce, Poland; (J.S.-K.); (S.G.)
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, IX Wieków Kielc 19A, 25-317 Kielce, Poland
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (P.M.); (E.G.)
| |
Collapse
|