1
|
Thierens NDE, Verdonk RC, Löhr JM, van Santvoort HC, Bouwense SA, van Hooft JE. Chronic pancreatitis. Lancet 2025; 404:2605-2618. [PMID: 39647500 DOI: 10.1016/s0140-6736(24)02187-1] [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: 04/19/2024] [Revised: 09/20/2024] [Accepted: 10/01/2024] [Indexed: 12/10/2024]
Abstract
Chronic pancreatitis is a progressive fibroinflammatory disease primarily caused by a complex interplay of environmental and genetic risk factors. It might result in pancreatic exocrine and endocrine insufficiency, chronic pain, reduced quality of life, and increased mortality. The diagnosis is based on the presence of typical symptoms and multiple morphological manifestations of the pancreas, including pancreatic duct stones and strictures, parenchymal calcifications, and pseudocysts. Management of chronic pancreatitis consists of prevention and treatment of complications, requiring a multidisciplinary approach focusing on lifestyle modifications, exocrine insufficiency, nutritional status, bone health, endocrine insufficiency, pain management, and psychological care. To optimise clinical outcomes, screening for complications and evaluation of treatment efficacy are indicated in all patients with chronic pancreatitis.
Collapse
Affiliation(s)
- Naomi DE Thierens
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, Netherlands; Department of Research and Development, St Antonius Hospital, Nieuwegein, Netherlands.
| | - Robert C Verdonk
- Department of Gastroenterology and Hepatology, St Antonius Hospital, Nieuwegein, Netherlands
| | - J Matthias Löhr
- Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Hjalmar C van Santvoort
- Department of Surgery, St Antonius Hospital, Nieuwegein, Netherlands; Department of Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Stefan Aw Bouwense
- Department of Surgery, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Jeanin E van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
2
|
Murray K, Oldfield L, Stefanova I, Gentiluomo M, Aretini P, O'Sullivan R, Greenhalf W, Paiella S, Aoki MN, Pastore A, Birch-Ford J, Rao BH, Uysal-Onganer P, Walsh CM, Hanna GB, Narang J, Sharma P, Campa D, Rizzato C, Turtoi A, Sever EA, Felici A, Sucularli C, Peduzzi G, Öz E, Sezerman OU, Van der Meer R, Thompson N, Costello E. Biomarkers, omics and artificial intelligence for early detection of pancreatic cancer. Semin Cancer Biol 2025; 111:76-88. [PMID: 39986585 DOI: 10.1016/j.semcancer.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is frequently diagnosed in its late stages when treatment options are limited. Unlike other common cancers, there are no population-wide screening programmes for PDAC. Thus, early disease detection, although urgently needed, remains elusive. Individuals in certain high-risk groups are, however, offered screening or surveillance. Here we explore advances in understanding high-risk groups for PDAC and efforts to implement biomarker-driven detection of PDAC in these groups. We review current approaches to early detection biomarker development and the use of artificial intelligence as applied to electronic health records (EHRs) and social media. Finally, we address the cost-effectiveness of applying biomarker strategies for early detection of PDAC.
Collapse
Affiliation(s)
- Kate Murray
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Lucy Oldfield
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Irena Stefanova
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | | | | | - Rachel O'Sullivan
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - William Greenhalf
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Salvatore Paiella
- Pancreatic Surgery Unit, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Italy
| | - Mateus N Aoki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Brazil
| | - Aldo Pastore
- Fondazione Pisana per la Scienza, Scuola Normale Superiore di Pisa, Italy
| | - James Birch-Ford
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Bhavana Hemantha Rao
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Czech Republic
| | - Pinar Uysal-Onganer
- School of Life Sciences, Cancer Mechanisms and Biomarkers Group, The University of Westminster, United Kingdom
| | - Caoimhe M Walsh
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - George B Hanna
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | | | | | | | | | - Andrei Turtoi
- Tumor Microenvironment and Resistance to Treatment Lab, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, France
| | - Elif Arik Sever
- Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | | | | | | | - Elif Öz
- Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | - Osman Uğur Sezerman
- Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Turkiye
| | | | | | - Eithne Costello
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom.
| |
Collapse
|
3
|
Huang C, Shen Y, Galgano SJ, Goenka AH, Hecht EM, Kambadakone A, Wang ZJ, Chu LC. Advancements in early detection of pancreatic cancer: the role of artificial intelligence and novel imaging techniques. Abdom Radiol (NY) 2025; 50:1731-1743. [PMID: 39467913 DOI: 10.1007/s00261-024-04644-7] [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: 08/15/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024]
Abstract
Early detection is crucial for improving survival rates of pancreatic ductal adenocarcinoma (PDA), yet current diagnostic methods can often fail at this stage. Recently, there has been significant interest in improving risk stratification and developing imaging biomarkers, through novel imaging techniques, and most notably, artificial intelligence (AI) technology. This review provides an overview of these advancements, with a focus on deep learning methods for early detection of PDA.
Collapse
Affiliation(s)
| | - Yiqiu Shen
- New York University Langone Health, New York, USA
| | | | | | | | | | - Zhen Jane Wang
- University of California, San Francisco, San Francisco, USA
| | - Linda C Chu
- Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
4
|
Engels MML, Berger CK, Mahoney DW, Hoogenboom SA, Sarwal D, Klatte DCF, De La Fuente J, Gandhi S, Taylor WR, Foote PH, Doering KA, Delgado AM, Burger KN, Abu Dayyeh BK, Bofill-Garcia A, Brahmbhatt B, Chandrasekhara V, Gleeson FC, Gomez V, Kumbhari V, Law RJ, Lukens FJ, Raimondo M, Rajan E, Storm AC, Vargas Valls EJ, van Hooft JE, Wallace MB, Kisiel JB, Majumder S. Multimodal Pancreatic Cancer Detection Using Methylated DNA Biomarkers in Pancreatic Juice and Plasma CA 19-9: A Prospective Multicenter Study. Clin Gastroenterol Hepatol 2025; 23:766-775. [PMID: 39477082 PMCID: PMC11930620 DOI: 10.1016/j.cgh.2024.07.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/14/2024] [Accepted: 07/30/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND AND AIMS In previous studies, methylated DNA markers (MDMs) have been identified in pancreatic juice (PJ) for detecting pancreatic ductal adenocarcinoma (PDAC). In this prospective multicenter study, the sensitivity and specificity characteristics of this panel of PJ-MDMs was evaluated standalone and in combination with plasma carbohydrate antigen 19-9 (CA 19-9). METHODS Paired PJ and plasma were assayed from 88 biopsy-proven treatment-naïve PDAC cases and 134 controls (53 with normal pancreas, 23 with chronic pancreatitis [CP], 58 with intraductal papillary mucinous neoplasm). Bisulfite-converted DNA from buffered PJ was analyzed using long-probe quantitative amplified signal assay targeting 14 MDMs (NDRG4, BMP3, TBX15, C13orf18, PRKCB, CLEC11A, CD1D, ELMO1, IGF2BP1, RYR2, ADCY1, FER1L4, EMX1, and LRRC4) and a reference gene (methylated B3GALT6). Logistic regression was used to fit the previously identified 3-MDM PJ panel (FER1L4, C13orf18, and BMP3). Discrimination accuracy was summarized using area under the receiver-operating characteristic curve (AUROC) with corresponding 95% confidence interval (CI). RESULTS Methylated FER1L4 had the highest individual AUROC of 0.83 (95% CI, 0.78-0.89). The AUROC for the 3-MDM PJ + plasma CA 19-9 model (0.95; 95% CI, 0.92-0.98) was higher than both the 3-MDM PJ panel (0.87; 95% CI, 0.82-0.92)) and plasma CA 19-9 alone (0.91; 95% CI, 0.87-0.96) (P = .0002 and .0135, respectively). At a specificity of 88% (95% CI, 81%-93%), the sensitivity of this model was 89% (95% CI, 80%-94%) for all PDAC stages and 83% (95% CI, 64%-94%) for stage I/II PDAC. CONCLUSIONS A panel combining PJ-MDMs and plasma CA 19-9 discriminates PDAC from both healthy and disease control groups with high accuracy. This provides support for combining PJ and blood-based biomarkers for enhancing diagnostic sensitivity and successful early PDAC detection.
Collapse
Affiliation(s)
- Megan M L Engels
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida; Division of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Calise K Berger
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Douglas W Mahoney
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, Minnesota
| | - Sanne A Hoogenboom
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida; Division of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dhruv Sarwal
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Derk C F Klatte
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida; Division of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jaime De La Fuente
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Sonal Gandhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - William R Taylor
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Patrick H Foote
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Karen A Doering
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Adriana M Delgado
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Kelli N Burger
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, Minnesota
| | - Barham K Abu Dayyeh
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - Bhaumik Brahmbhatt
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | | | - Ferga C Gleeson
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Victoria Gomez
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | - Vivek Kumbhari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | - Ryan J Law
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Frank J Lukens
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | - Massimo Raimondo
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | - Elizabeth Rajan
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Andrew C Storm
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Eric J Vargas Valls
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Jeanin E van Hooft
- Division of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Michael B Wallace
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida; Division of Gastroenterology and Hepatology, Sheikh Shakhbout Medical City, Abu Dhabi, United Arab Emirates
| | - John B Kisiel
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
5
|
Xu K, Wang X, Zhou C, Zuo J, Zeng C, Zhou P, Zhang L, Gao X, Wang X. Synergic value of 3D CT-derived body composition and triglyceride glucose body mass for survival prognostic modeling in unresectable pancreatic cancer. Front Nutr 2025; 12:1499188. [PMID: 40177184 PMCID: PMC11961436 DOI: 10.3389/fnut.2025.1499188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Background Personalized and accurate survival risk prognostication remains a significant challenge in advanced pancreatic ductal adenocarcinoma (PDAC), despite extensive research on prognostic and predictive markers. Patients with PDAC are prone to muscle loss, fat consumption, and malnutrition, which is associated with inferior outcomes. This study investigated the use of three-dimensional (3D) anthropometric parameters derived from computed tomography (CT) scans and triglyceride glucose-body mass index (TyG-BMI) in relation to overall survival (OS) outcomes in advanced PDAC patients. Additionally, a predictive model for 1 year OS was developed based on body components and hematological indicators. Methods A retrospective analysis was conducted on 303 patients with locally advanced PDAC or synchronous metastases undergoing first-line chemotherapy, all of whom had undergone pretreatment abdomen-pelvis CT scans. Automatic 3D measurements of subcutaneous and visceral fat volume, skeletal muscle volume, and skeletal muscle density (SMD) were assessed at the L3 vertebral level by an artificial intelligence assisted diagnosis system (HY Medical). Various indicators including TyG-BMI, nutritional indicators [geriatric nutritional risk index (GNRI) and prealbumin], and inflammation indicators [(C-reactive protein (CRP) and neutrophil to lymphocyte ratio (NLR)] were also recorded. All patients underwent follow-up for at least 1 year and a dynamic nomogram for personalized survival prediction was constructed. Results We included 211 advanced PDAC patients [mean (standard deviation) age, 63.4 ± 11.2 years; 89 women (42.2) %)]. Factors such as low skeletal muscle index (SMI) (P = 0.011), high visceral to subcutaneous adipose tissue area ratio (VSR) (P < 0.001), high visceral fat index (VFI) (P < 0.001), low TyG-BMI (P = 0.004), and low prealbumin (P = 0.001) were identified as independent risk factors associated with 1 year OS. The area under the curve of the established dynamic nomogram was 0.846 and the calibration curve showed good consistency. High-risk patients (> 211.9 points calculated using the nomogram) had significantly reduced survival rates. Conclusion In this study, the proposed nomogram model (with web-based tool) enabled individualized prognostication of OS and could help to guide risk-adapted nutritional treatment for patients with unresectable PDAC or synchronous metastases.
Collapse
Affiliation(s)
- Kangjing Xu
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinbo Wang
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Changsheng Zhou
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Junbo Zuo
- Department of General Surgery, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Chenghao Zeng
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Pinwen Zhou
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Li Zhang
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xuejin Gao
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinying Wang
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
6
|
Matsubayashi H, Kiyozumi Y, Ono H. Genetic medicine of familial and hereditary pancreatic cancer: Recent update in the era of precision cancer medicine. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2025; 32:212-227. [PMID: 39814596 DOI: 10.1002/jhbp.12112] [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: 01/18/2025]
Abstract
In Japan, 5 years have passed since the initiation of precision cancer medicine, and recent data accumulation in familial pancreatic cancer (FPC) and hereditary pancreatic cancer is outstanding. Multigene germline panel tests (MGPTs) have revealed that 7%-18% of patients with pancreatic cancer (PC) harbor pathogenic germline variants (PGVs), almost equal to the levels of breast, ovarian, endometrial, and colorectal cancers, with a higher incidence in FPC (14%-26%). The majority of PGVs seen in PC patients are clinically actionable and associated with homologous recombination (HR) pathways (6%-10%, particularly BRCA1/2 in 5%-6%), and the clinical guidelines recommend or propose genetic testing for all PC patients. Consensus guidelines have been established for most of the hereditary syndromes associated with PC risks, and surveillances of the pancreas and other at-risk organs are recommended for PGV carriers. Hereditary breast and ovarian cancer (HBOC) is the commonest hereditary cancer syndrome that has moderately increasing life-time risks of PC (3%-7% in Western countries); however, recent Japanese research demonstrated a higher risk level (BRCA1: 16%, BRCA2: 14%). Moreover, recent evidence has suggested a risk linkage between PC and ovarian cancer in HBOC pedigrees. High scores of homologous recombination deficiency suggest biallelic dysfunction of BRCA or other HR-related genes, and the likely effectiveness of platinum agents and PARP inhibitors against PCs. Remote counseling and testing are possible option in the future genetic medicine. As PC ranks in the second commonest target of precision cancer medicine in Japan, we must treat the patients and manage their at-risk relatives efficiently.
Collapse
Affiliation(s)
- Hiroyuki Matsubayashi
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, Shizuoka, Japan
- Division of Endoscopy, Shizuoka Cancer Center, Shizuoka, Japan
| | - Yoshimi Kiyozumi
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, Shizuoka, Japan
| | - Hiroyuki Ono
- Division of Endoscopy, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
7
|
Vanek P, Freeman ML. Updates in the Diagnosis of Chronic Pancreatitis: Current Approaches and New Possibilities. Gastroenterol Clin North Am 2025; 54:143-156. [PMID: 39880524 DOI: 10.1016/j.gtc.2024.08.007] [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
This review provides a comprehensive update on the diagnostic approaches to chronic pancreatitis (CP), emphasizing recent advancements in imaging techniques, biomarker research, and multivariable scoring systems. Despite substantial progress in these areas, current diagnostic algorithms have limitations, particularly for early and non-calcific CP. Traditional criteria have focused on classic diagnostic signs, but "minimal change" CP is increasingly recognized through advanced imaging and function tests. This article aims to guide clinicians in applying current methods and available strategies for CP diagnosis and outline research efforts in the field.
Collapse
Affiliation(s)
- Petr Vanek
- Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 77900 Olomouc, Czech Republic; Department of Gastroenterology and Digestive Endoscopy, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 65653 Brno, Czech Republic
| | - Martin L Freeman
- Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota, 420 Delaware Street Southeast, Minneapolis, MN 55455, USA.
| |
Collapse
|
8
|
Desolneux G, Castanet F. [Precancerous lesions of the pancreas, prophylactic surgery]. Bull Cancer 2025; 112:263-269. [PMID: 40049795 DOI: 10.1016/j.bulcan.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 03/15/2025]
Abstract
Pancreatic cancer is a serious pathology whose incidence is increasing. Advances in imaging have made it possible to fortuitously highlight more and more cystic pancreatic lesions, some of which have a malignant potential, leading to the consideration of prophylactic excision. The various known precancerous lesions of the pancreas as well as current management recommendations are here reviewed.
Collapse
Affiliation(s)
- Gregoire Desolneux
- Chirurgie digestive oncologique, institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux cedex, France.
| | - Fanny Castanet
- Chirurgie digestive oncologique, institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux cedex, France
| |
Collapse
|
9
|
Afghani E, Lennon AM. What Is the Latest in Pancreatic Cysts? Gastroenterol Clin North Am 2025; 54:189-203. [PMID: 39880527 DOI: 10.1016/j.gtc.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Pancreatic cysts are common incidental findings. The understanding of pancreatic cysts has evolved tremendously over the past few decades. Molecular diagnostic and endoscopic techniques have led to more precise characterization of cyst types and interventions to improve patient outcomes. This article outlines these recent innovations in pancreatic cyst diagnosis and management.
Collapse
Affiliation(s)
- Elham Afghani
- Department of Medicine, Johns Hopkins University, 1830 East Monument Street, Room 436, Baltimore, MD 21287, USA
| | - Anne Marie Lennon
- Department of Medicine, University of Pittsburgh, 3550 Terrace Street, 1218 Scaife Hall, Pittsburgh, PA 15261, USA.
| |
Collapse
|
10
|
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
|
11
|
Donadio MDS, de Jesus VHF. Stepped-wedge clinical trials in pancreatic cancer: a step backward in improving overall survival or a leap forward to enhance quality of care? Gland Surg 2025; 14:263-267. [PMID: 40115846 PMCID: PMC11921292 DOI: 10.21037/gs-24-472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/03/2025] [Indexed: 03/23/2025]
Affiliation(s)
| | - Victor Hugo Fonseca de Jesus
- Department of Gastrointestinal Medical Oncology, Oncoclínicas, Florianópolis, SC, Brazil
- Department of Medical Oncology, Centro de Pesquisas Oncológicas (CEPON), Florianópolis, SC, Brazil
| |
Collapse
|
12
|
Renjifo-Correa ME, Fanni SC, Bustamante-Cristancho LA, Cuibari ME, Aghakhanyan G, Faggioni L, Neri E, Cioni D. Diagnostic Accuracy of Radiomics in the Early Detection of Pancreatic Cancer: A Systematic Review and Qualitative Assessment Using the Methodological Radiomics Score (METRICS). Cancers (Basel) 2025; 17:803. [PMID: 40075651 PMCID: PMC11898638 DOI: 10.3390/cancers17050803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) is an aggressive and lethal malignancy with increasing incidence and low survival rate, primarily due to the late detection of the disease. Radiomics has demonstrated its utility in recognizing patterns and anomalies not perceptible to the human eye. This systematic literature review aims to assess the application of radiomics in the analysis of pancreatic parenchyma images to identify early indicators predictive of PDAC. METHODS A systematic search of original research papers was performed on three databases: PubMed, Embase, and Scopus. Two reviewers applied the inclusion and exclusion criteria, and one expert solved conflicts for selecting the articles. After extraction and analysis of the data, there was a quality assessment of these articles using the Methodological Radiomics Score (METRICS) tool. The METRICS assessment was carried out by two raters, and conflicts were solved by a third reviewer. RESULTS Ten articles for analysis were retrieved. CT scan was the diagnostic imaging used in all the articles. All the studies were retrospective and published between 2019 and 2024. The main objective of the articles was to generate radiomics-based machine learning models able to differentiate pancreatic tumors from healthy tissue. The reported diagnostic performance of the model chosen yielded very high results, with a diagnostic accuracy between 86.5% and 99.2%. Texture and shape features were the most frequently implemented. The METRICS scoring assessment demonstrated that three articles obtained a moderate quality, five a good quality, and, finally, two articles yielded excellent quality. The lack of external validation and available model, code, and data were the major limitations according to the qualitative assessment. CONCLUSIONS There is high heterogeneity in the research question regarding radiomics and pancreatic cancer. The principal limitations of the studies were mainly due to the nature of the trials and the considerable heterogeneity of the radiomic features reported. Nonetheless, the work in this field is promising, and further studies are still required to adopt radiomics in the early detection of PDAC.
Collapse
Affiliation(s)
- María Estefanía Renjifo-Correa
- Radiology Department, Magnetic Resonance Service, Clínica de Occidente, Calle 18 Norte No. 5N 34, Cali 760045, Colombia;
| | - Salvatore Claudio Fanni
- Department of Translational Research, Academic Radiology, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (M.E.C.); (G.A.); (L.F.); (E.N.); (D.C.)
| | | | - Maria Emanuela Cuibari
- Department of Translational Research, Academic Radiology, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (M.E.C.); (G.A.); (L.F.); (E.N.); (D.C.)
| | - Gayane Aghakhanyan
- Department of Translational Research, Academic Radiology, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (M.E.C.); (G.A.); (L.F.); (E.N.); (D.C.)
| | - Lorenzo Faggioni
- Department of Translational Research, Academic Radiology, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (M.E.C.); (G.A.); (L.F.); (E.N.); (D.C.)
| | - Emanuele Neri
- Department of Translational Research, Academic Radiology, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (M.E.C.); (G.A.); (L.F.); (E.N.); (D.C.)
| | - Dania Cioni
- Department of Translational Research, Academic Radiology, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (M.E.C.); (G.A.); (L.F.); (E.N.); (D.C.)
| |
Collapse
|
13
|
Wang S, Zhou H, Cai K, Fan Y, Yang X, Zhang B, Wu Y. Predictive value of perioperative fasting blood glucose for post pancreatectomy diabetes mellitus in pancreatic ductal carcinoma patients. World J Surg Oncol 2025; 23:55. [PMID: 39955538 PMCID: PMC11830169 DOI: 10.1186/s12957-025-03705-5] [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: 10/23/2024] [Accepted: 02/04/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND To explore the risk factors of post pancreatectomy diabetes mellitus (PPTDM)in pancreatic ductal carcinoma (PDAC) patients and the value of perioperative fasting blood glucose (FBG) level expression on the long-term survival after surgery. MATERIALS AND METHODS Between December 2015 and December 2019, a cohort of 509 patients diagnosed with PDAC and undergoing resection at our hospital was analyzed. They were stratified into two groups, Control group (Control) and study group (PPTDM), depending on the onset of postoperative diabetes mellitus. We analyzed the survival rates at 6 months, 12 months and 24 months post-operation in the two groups. We use univariate and logostic multivariate regressions to analyze the risk factors for PPTDM. ROC curve analysis was conducted to assess the diagnostic significance of perioperative FBG levels regarding patients' long-term survival rates. The Kaplan-Meier method was employed to assess the impact of both preoperative and postoperative FBG levels on the survival rates within 24 months for each patient group. RESULTS The comparison of general clinical data between the two groups shows marginal differences without statistical significance(P > 0.05); Patients in PPTDM group had significantly higher BMI, preoperative jaundice proportion, larger tumor diameter, higher TNM stage and higher proportion of distal pancreatectomy (DP), with P values of 0.023, 0.010, 0.040, 0.012 and 0.005, respectively. The levels of preoperative FBG and postoperative FBG in PPTDM patients exhibited statistically significant elevation compared to the control group (P < 0.05). There were no significant differences in surgery-related indicators between the two groups in operative time, number of dissected positive lymph nodes, total number of dissected lymph nodes, intraoperative blood loss and other related data (P > 0.05). Hospitalization duration of PPTDM patients was longer than control group (P = 0.047). PPTDM group had significantly higher expression concentrations of BUN, Cr, TG, LDL and Apo-B factors (P = 0.023, 0.024, 0.013, 0.045 and 0.017). 17 patients (5.03%) died in the PPTDM group and 4 patients (2.35%) in control group which had significantly difference (P = 0.020). In univariate and logostic multivariate regression analysis indicated tumor size, jaundice, BUN, Cr, TG, LDL, Apo-B concentrations and DP approach were significantly correlated to the risk for PPTDM (P < 0.05). ROC curve analysis results showed combining of preoperative and postoperation FBG showed the highest diagnostic efficacy, followed by postoperation FBG and preoperative FBG. The AUC areas of the three groups were 0.745, 0.623 and 0.588, respectively, and the critical values of the three groups were 9.81/9.95 mmol/L, 10.18 mmol/L and 10.23 mmol/L, respectively, with statistical significance (P < 0.05). Results were considered statistically significant if the p-value was less than 0.05. CONCLUSION PPTDM stands as a significant postoperative complication following pancreatic cancer surgery, characterized by a high incidence and severity. Several risk factors have garnered considerable attention among clinical surgeon. PPTDM may be an influential factor in postoperative prognosis of pancreatic cancer. The expression levels of preoperative and postoperative blood glucose hold diagnostic value for the long-term prognosis of pancreatic cancer patients. Early regulation and intervention by surgeons concerning perioperative FBG could potentially mitigate the risk of PPTDM.
Collapse
Affiliation(s)
- Shuai Wang
- Department of General Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, N1 Shangcheng Road, Yiwu, Zhejiang, 322000, China
| | - Hanshen Zhou
- Department of General Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, N1 Shangcheng Road, Yiwu, Zhejiang, 322000, China
| | - Kaili Cai
- Department of General Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, N1 Shangcheng Road, Yiwu, Zhejiang, 322000, China
| | - Yiqun Fan
- Department of Hepato-Pancreato-Biliary Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou, 310009, China
| | - Xiaohui Yang
- Department of General Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, N1 Shangcheng Road, Yiwu, Zhejiang, 322000, China
| | - Bo Zhang
- Department of Hepato-Pancreato-Biliary Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou, 310009, China
| | - Yulian Wu
- Department of Hepato-Pancreato-Biliary Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Shangcheng District, Hangzhou, 310009, China.
- Department of General Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, N1 Shangcheng Road, Yiwu, Zhejiang, 322000, China.
| |
Collapse
|
14
|
Kimura H, Lahouel K, Tomasetti C, Roberts NJ. Functional characterization of all CDKN2A missense variants and comparison to in silico models of pathogenicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.28.573507. [PMID: 38234851 PMCID: PMC10793438 DOI: 10.1101/2023.12.28.573507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Interpretation of variants identified during genetic testing is a significant clinical challenge. In this study, we developed a high-throughput CDKN2A functional assay and characterized all possible CDKN2A missense variants. We found that 17.7% of all missense variants were functionally deleterious. We also used our functional classifications to assess the performance of in silico models that predict the effect of variants, including recently reported models based on machine learning. Notably, we found that all in silico models performed similarly when compared to our functional classifications with accuracies of 39.5-85.4%. Furthermore, while we found that functionally deleterious variants were enriched within ankyrin repeats, we did not identify any residues where all missense variants were functionally deleterious. Our functional classifications are a resource to aid the interpretation of CDKN2A variants and have important implications for the application of variant interpretation guidelines, particularly the use of in silico models for clinical variant interpretation.
Collapse
Affiliation(s)
- Hirokazu Kimura
- Department of Pathology, the Johns Hopkins University School of Medicine; Baltimore, 21287, USA
| | - Kamel Lahouel
- Division of Integrated Genomics, Translational Genomics Research Institute; Phoenix, 85004, USA
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope; Duarte, 91010, USA
| | - Cristian Tomasetti
- Division of Integrated Genomics, Translational Genomics Research Institute; Phoenix, 85004, USA
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope; Duarte, 91010, USA
| | - Nicholas J. Roberts
- Department of Pathology, the Johns Hopkins University School of Medicine; Baltimore, 21287, USA
- Department of Oncology, the Johns Hopkins University School of Medicine; Baltimore, 21287, USA
| |
Collapse
|
15
|
Heller M, Mann DA, Katona BW. Current Approaches of Pancreatic Cancer Surveillance in High-Risk Individuals. J Gastrointest Cancer 2025; 56:61. [PMID: 39932614 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
|
16
|
Jin D, Khan NU, Gu W, Lei H, Goel A, Chen T. Informatics strategies for early detection and risk mitigation in pancreatic cancer patients. Neoplasia 2025; 60:101129. [PMID: 39842383 PMCID: PMC11763847 DOI: 10.1016/j.neo.2025.101129] [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: 11/07/2024] [Revised: 01/17/2025] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
This review provides a comprehensive overview of the current landscape in pancreatic cancer (PC) screening, diagnosis, and early detection. This emphasizes the need for targeted screening in high-risk groups, particularly those with familial predispositions and genetic mutations, such as BRCA1, BRCA2, and PALB2. This review highlights the sporadic nature of most PC cases and significant risk factors, including smoking, alcohol consumption, obesity, and diabetes. Advanced imaging techniques, such as Endoscopic Ultrasound (EUS) and Contrast-Enhanced Harmonic Imaging (CEH-EUS), have been discussed for their superior sensitivity in early detection. This review also explores the potential of novel biomarkers, including those found in body fluids, such as serum, plasma, urine, and bile, as well as the emerging role of liquid biopsy technologies in analyzing circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and exosomes. AI-driven approaches, such as those employed in Project Felix and CancerSEEK, have been highlighted for their potential to enhance early detection through deep learning and biomarker discovery. This review underscores the importance of universal genetic testing and the integration of AI with traditional diagnostic methods to improve outcomes in high-risk individuals. Additionally, this review points to future directions in PC diagnostics, including next-generation imaging, molecular biomarkers, and personalized medicine, aiming to overcome current diagnostic challenges and improve survival rates. Ultimately, the review advocates the adoption of informatics and AI-driven strategies to enhance early detection, reduce morbidity, and save lives in the fight against pancreatic cancer.
Collapse
Affiliation(s)
- Di Jin
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, China; Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Najeeb Ullah Khan
- Institute of Biotechnology & Genetic Engineering (Health Division), The University of Agriculture Peshawar, Peshawar, PO Box 25130, Pakistan.
| | - Wei Gu
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, China; Wenzhou Medical University, Wenzhou, 325000, China.
| | - Huijun Lei
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, China.
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California, USA; City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| | - Tianhui Chen
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310018, China.
| |
Collapse
|
17
|
Solaiyappan M, Bharti SK, Sharma RK, Dbouk M, Nizam W, Brock MV, Goggins MG, Bhujwalla ZM. Artificial neural network detection of pancreatic cancer from proton (1H) magnetic resonance spectroscopy patterns of plasma metabolites. COMMUNICATIONS MEDICINE 2025; 5:24. [PMID: 39838068 PMCID: PMC11751387 DOI: 10.1038/s43856-024-00727-0] [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: 02/15/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Routine screening to detect silent but deadly cancers such as pancreatic ductal adenocarcinoma (PDAC) can significantly improve survival, creating an important need for a convenient screening test. High-resolution proton (1H) magnetic resonance spectroscopy (MRS) of plasma identifies circulating metabolites that can allow detection of cancers such as PDAC that have highly dysregulated metabolism. METHODS We first acquired 1H MR spectra of human plasma samples classified as normal, benign pancreatic disease and malignant (PDAC). We next trained a system of artificial neural networks (ANNs) to process and discriminate these three classes using the full spectrum range and resolution of the acquired spectral data. We then identified and ranked spectral regions that played a salient role in the discrimination to provide interpretability of the results. We tested the accuracy of the ANN performance using blinded plasma samples. RESULTS We show that our ANN approach yields, in a cross validation-based training of 170 samples, a sensitivity and a specificity of 100% for malignant versus non-malignant (normal and disease combined) discrimination. The trained ANNs achieve a sensitivity and specificity of 87.5% and 93.1% respectively (AUC: ROC = 0.931, P-R = 0.854), with 45 blinded plasma samples. Further, we show that the salient spectral regions of the ANN discrimination correspond to metabolites of known importance for their role in cancers. CONCLUSIONS Our results demonstrate that the ANN approach presented here can identify PDAC from 1H MR plasma spectra to provide a convenient plasma-based assay for population-level screening of PDAC. The ANN approach can be suitably expanded to detect other cancers with metabolic dysregulation.
Collapse
Affiliation(s)
- Meiyappan Solaiyappan
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Santosh Kumar Bharti
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Raj Kumar Sharma
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mohamad Dbouk
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wasay Nizam
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Malcolm V Brock
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael G Goggins
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zaver M Bhujwalla
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
18
|
Zhan T, Betge J, Schulte N, Dreikhausen L, Hirth M, Li M, Weidner P, Leipertz A, Teufel A, Ebert MP. Digestive cancers: mechanisms, therapeutics and management. Signal Transduct Target Ther 2025; 10:24. [PMID: 39809756 PMCID: PMC11733248 DOI: 10.1038/s41392-024-02097-4] [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: 06/29/2024] [Revised: 10/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Cancers of the digestive system are major contributors to global cancer-associated morbidity and mortality, accounting for 35% of annual cases of cancer deaths. The etiologies, molecular features, and therapeutic management of these cancer entities are highly heterogeneous and complex. Over the last decade, genomic and functional studies have provided unprecedented insights into the biology of digestive cancers, identifying genetic drivers of tumor progression and key interaction points of tumor cells with the immune system. This knowledge is continuously translated into novel treatment concepts and targets, which are dynamically reshaping the therapeutic landscape of these tumors. In this review, we provide a concise overview of the etiology and molecular pathology of the six most common cancers of the digestive system, including esophageal, gastric, biliary tract, pancreatic, hepatocellular, and colorectal cancers. We comprehensively describe the current stage-dependent pharmacological management of these malignancies, including chemo-, targeted, and immunotherapy. For each cancer entity, we provide an overview of recent therapeutic advancements and research progress. Finally, we describe how novel insights into tumor heterogeneity and immune evasion deepen our understanding of therapy resistance and provide an outlook on innovative therapeutic strategies that will shape the future management of digestive cancers, including CAR-T cell therapy, novel antibody-drug conjugates and targeted therapies.
Collapse
Affiliation(s)
- Tianzuo Zhan
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johannes Betge
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Schulte
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Dreikhausen
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Hirth
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Moying Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philip Weidner
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Antonia Leipertz
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Teufel
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany.
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
19
|
Cox M, Vitello DJ, Chawla A. The Current Role of Circulating Tumor DNA in the Management of Pancreatic Cancer. J Gastrointest Cancer 2025; 56:44. [PMID: 39808248 DOI: 10.1007/s12029-024-01129-0] [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] [Accepted: 10/23/2024] [Indexed: 01/16/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is projected to be the second leading cause of cancer-related death by 2030. Early identification is rare, with a 5-year overall survival (OS) of less than 10%. Advances in the understanding of PDAC tumor biology are needed to improve these outcomes. Circulating tumor DNA (ctDNA) represents a promising novel biomarker in the identification and management of PDAC. Drawn from peripheral blood and analyzed using a variety of techniques, the detection of ctDNA in PDAC has been associated with shorter OS, minimal residual disease presence, and shorter recurrence-free survival. The use of ctDNA has also been examined as an indicator of therapeutic resistance, susceptibility to targeted therapy, and therapeutic response. While promising, ctDNA analysis is limited by its low rates of detection in some settings and lack of predictive ability in others. Many studies examining the utility of ctDNA for the management of PDAC have been relatively small retrospective cohort studies. The current findings will need to be validated by incorporation of ctDNA analysis into cancer registries and larger prospective studies. Given the current, rapid evolution in the field, it is possible that with time, ctDNA will be more routinely incorporated into the clinical management of PDAC.
Collapse
Affiliation(s)
- Madison Cox
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, USA
| | - Dominic J Vitello
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Akhil Chawla
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, USA.
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
20
|
Blanco Abad C, Gomila Pons P, Campos Ramírez S, Álvarez Alejandro M, Torres Ramón MI, Miramar Gallart MD, Izquierdo Álvarez S, Polo Marques E, Pazo Cid R. Hereditary Pancreatic Cancer: Advances in Genetic Testing, Early Detection Strategies, and Personalized Management. J Clin Med 2025; 14:367. [PMID: 39860372 PMCID: PMC11766428 DOI: 10.3390/jcm14020367] [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: 11/14/2024] [Revised: 12/29/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with a five-year survival rate of approximately 13% for advanced stages. While the majority of PDAC cases are sporadic, a significant subset is attributable to hereditary and familial predispositions, accounting for approximately 25% of cases. This article synthesizes recent advancements in the understanding, detection, and management of hereditary pancreatic cancer (PC). Results: Our review highlights the critical role of genetic testing (GT) in identifying high-risk individuals (HRIs), with germline pathogenic variants (PVs) found in up to 20% of hereditary PDAC cases. Since the implementation of next-generation sequencing (NGS) panels in 2014, detection capabilities have been significantly enhanced. HRIs can be included in screening programs that facilitate the early detection of PDAC. Early detection strategies, including the use of microribonucleic acid (miRNAs) signatures and novel imaging techniques like hyperpolarized 13C-magnetic resonance spectroscopy (MRS) have shown promising results. The identification of germline pathogenic variants (PVs) or mutations in homologous recombination (HR) genes plays a predictive role in the response to various treatments, prolonging patient survival. Discussion: Universal germline testing for PDAC, as recommended by the National Comprehensive Cancer Network (NCCN), is now a standard practice, facilitating the identification of at-risk individuals and enabling targeted surveillance and intervention. Multidisciplinary management, integrating genetic counseling, imaging, and gastrointestinal services, is essential for optimizing outcomes. Conclusions: Advances in genetic testing and biomarker research are transforming the landscape of hereditary PC management. Early detection and personalized treatment strategies are pivotal in improving survival rates. Ongoing multi-institutional research efforts are crucial for validating biomarkers and developing preventive measures, ultimately aiming to reduce the burden of this aggressive cancer.
Collapse
Affiliation(s)
- Carmen Blanco Abad
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
| | - Paula Gomila Pons
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
| | - Sara Campos Ramírez
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
| | - María Álvarez Alejandro
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
- Medical Oncology Department, Hospital Clinico Universitario Lozano Blesa, 50009 Zaragoza, Spain
| | - María Irene Torres Ramón
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
- Medical Oncology Department, Hospital Clinico Universitario Lozano Blesa, 50009 Zaragoza, Spain
| | | | - Silvia Izquierdo Álvarez
- Genetics Unit, Biochemistry Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
| | - Eduardo Polo Marques
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
- Aragon Institute of Health Sciences (IIS-A), 50012 Zaragoza, Spain
| | - Roberto Pazo Cid
- Medical Oncology Department, Hospital Universitario Miguel Servet, 50012 Zaragoza, Spain
- Medical Oncology Department, Hospital Clinico Universitario Lozano Blesa, 50009 Zaragoza, Spain
- Department of Medicine, Psychiatry and Dermatology, Faculty of Medicine, Zaragoza University, 50009 Zaragoza, Spain
| |
Collapse
|
21
|
Kryklyva V, Pflüger MJ, Ouchene H, Volleberg-Gorissen H, Mensenkamp AR, Jonker MA, van de Water C, Nagtegaal ID, Ligtenberg MJL, Brosens LAA. Germline Pathogenic Variants in Patients with Pancreatic Ductal Adenocarcinoma and Extra-Pancreatic Malignancies: A Nationwide Database Analysis. Mod Pathol 2025; 38:100709. [PMID: 39793706 DOI: 10.1016/j.modpat.2025.100709] [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: 05/21/2024] [Revised: 11/11/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease. Approximately 10% of affected individuals have an inherited component. Deleterious germline variants increase the lifetime risk for PDAC and are often associated with an elevated risk for extra-pancreatic malignancies. In this study, we aimed to determine the prevalence and impact of germline pathogenic variants (gPVs) in patients with PDAC and extra-pancreatic malignancies. Using tissue samples and longitudinal data from a nationwide pathology database, we identified patients with PDAC and a set of 7 extra-pancreatic malignancies to investigate the presence of gPVs in 25 cancer susceptibility genes with targeted next-generation sequencing. Of 473 patients with PDAC and at least 1 extra-pancreatic malignancy, 75 (16%) had gPVs. These were predominantly in ATM (n = 22), CDKN2A (n = 14), BRCA2 (n = 10), or CHEK2 (n = 10) genes. The combination of PDAC and ovarian carcinoma carried the highest prevalence of gPVs (4 of 10; 40%), followed by PDAC and melanoma (15 of 53; 28%), and PDAC and gastric cancer (2 of 9; 22%). Patients with PDAC and certain extra-pancreatic malignancies carry a higher burden of gPVs than unselected PDAC cohorts. This is a group that very likely benefits from genetic testing because germline status can have important diagnostic and therapeutic implications for affected individuals and their family members.
Collapse
Affiliation(s)
- Valentyna Kryklyva
- Department of Pathology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michael J Pflüger
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Surgery CCM|CVK, Charité - Universitätsmedizin Berlin, Germany; Department of Pathology, Graduate School of Life Sciences, Utrecht University, Utrecht, The Netherlands
| | - Hicham Ouchene
- Department of Human Genetics, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hanneke Volleberg-Gorissen
- Department of Human Genetics, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arjen R Mensenkamp
- Department of Human Genetics, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marianne A Jonker
- Department for Health Evidence, Section Biostatistics, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carlijn van de Water
- Department of Pathology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Iris D Nagtegaal
- Department of Pathology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Marjolijn J L Ligtenberg
- Department of Pathology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Human Genetics, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lodewijk A A Brosens
- Department of Pathology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
22
|
Mittendorf KF, Bland HT, Andujar J, Celaya-Cobbs N, Edwards C, Gerhart M, Hooker G, Hubert M, Jones SH, Marshall DR, Myers RA, Pratap S, Rosenbloom ST, Sadeghpour A, Wu RR, Orlando LA, Wiesner GL. Family history and cancer risk study (FOREST): A clinical trial assessing electronic patient-directed family history input for identifying patients at risk of hereditary cancer. Contemp Clin Trials 2025; 148:107714. [PMID: 39395532 DOI: 10.1016/j.cct.2024.107714] [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: 07/12/2024] [Revised: 09/16/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Hereditary cancer syndromes cause a high lifetime risk of early, aggressive cancers. Early recognition of individuals at risk can allow risk-reducing interventions that improve morbidity and mortality. Family health history applications that gather data directly from patients could alleviate barriers to risk assessment in the clinical appointment, such as lack of provider knowledge of genetics guidelines and limited time in the clinical appointment. New approaches allow linking these applications to patient health portals and their electronic health records (EHRs), offering an end-to-end solution for patient-input family history information and risk result clinical decision support for their provider. METHODS We describe the design of the first large-scale evaluation of an EHR-integrable, patient-facing family history software platform based on the Substitutable Medical Applications and Reusable Technologies on Fast Healthcare Interoperability Resources (SMART on FHIR) standard. In our study, we leverage an established implementation science framework to evaluate the success of our model to facilitate scalable, systematic risk assessment for hereditary cancers in diverse clinical environments in a large pragmatic study at two sites. We will also evaluate the success of the approach to improve the efficiency of downstream genetic counseling resulting from pre-counseling pedigree generation. CONCLUSIONS Our research study will provide evidence regarding a new care delivery model that is scalable and sustainable for a variety of medical centers and clinics. TRIAL REGISTRATION This study was registered on ClinicalTrials.gov under NCT05079334 on 15 October 2021.
Collapse
Affiliation(s)
- Kathleen F Mittendorf
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Harris T Bland
- Department of Biomedical Informatics, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Justin Andujar
- Division of Genetic Medicine, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Natasha Celaya-Cobbs
- Division of Genetic Medicine, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Clasherrol Edwards
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Meredith Gerhart
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gillian Hooker
- Division of Genetic Medicine, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA; Concert Genetics, Nashville, TN, USA
| | - Mryia Hubert
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sarah H Jones
- Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dana R Marshall
- Department of Pathology, Anatomy and Cell Biology, Meharry Medical College, Nashville, TN, USA
| | - Rachel A Myers
- Department of Medicine Clinical Research Unit, Duke University School of Medicine, Durham, NC, 27705, USA
| | - Siddharth Pratap
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - S Trent Rosenbloom
- Department of Biomedical Informatics, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Azita Sadeghpour
- Duke Precision Medicine Program, Department of Medicine, Duke University, Durham, NC, USA
| | - R Ryanne Wu
- Duke Precision Medicine Program, Department of Medicine, Duke University, Durham, NC, USA; 23andMe, Sunnyvale, CA, USA
| | - Lori A Orlando
- Duke Precision Medicine Program, Department of Medicine, Duke University, Durham, NC, USA
| | - Georgia L Wiesner
- Division of Genetic Medicine, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
23
|
Pal T, Schon KR, Astiazaran-Symonds E, Balmaña J, Foulkes WD, James P, Klugman S, Livinski AA, Mak JS, Ngeow J, Voian N, Wick MJ, Hanson H, Stewart DR, Tischkowitz M. Management of individuals with heterozygous germline pathogenic variants in ATM: A clinical practice resource of the American College of Medical Genetics and Genomics (ACMG). Genet Med 2025; 27:101243. [PMID: 39636577 DOI: 10.1016/j.gim.2024.101243] [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: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 12/07/2024] Open
Abstract
PURPOSE ATM germline pathogenic variants (GPVs) are associated with a moderately increased risk of female breast cancer, pancreatic cancer, and prostate cancer. Resources for managing ATM heterozygotes in clinical practice are limited. METHODS An international workgroup developed a clinical practice resource to guide management of ATM heterozygotes using peer-reviewed publications and expert opinion. RESULTS Although ATM is a moderate (intermediate) penetrance gene, cancer risks may be considered as a continuous variable, influenced by family history and other modifiers. ATM GPV heterozygotes should generally be offered enhanced breast surveillance according to their personalized risk estimate and country-specific guidelines and, generally, risk-reducing mastectomy is not recommended. Prostate cancer surveillance should be considered. Pancreatic cancer surveillance should be considered based on assessment of family history, ideally as part of a clinical trial, with existence of country-specific guidelines. For ATM GPV heterozygotes who develop cancer, radiation therapy decisions should not be influenced by the genetic result. Although poly-adenosine diphosphate ribose polymerase inhibitors are licensed for use in metastatic castration-resistant prostate cancer and ATM GPVs, the evidence-base is currently weak. CONCLUSION Systematic prospective data collection is needed to establish the spectrum of ATM-associated cancer and determine the outlines of surveillance, response to cancer treatment, and survival.
Collapse
Affiliation(s)
- Tuya Pal
- Department of Medicine, Vanderbilt University Medical Center/Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Katherine R Schon
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | | | - Judith Balmaña
- Hereditary Cancer Genetics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Medical Oncology Department, Hospital Universitari Vall d'Hebron, Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - William D Foulkes
- Departments of Human Genetics, Oncology and Medicine, McGill University, Montréal, Québec, Canada
| | - Paul James
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia; Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Susan Klugman
- Division of Reproductive & Medical Genetics, Department of Obstetrics and Gynecology and Women's Health, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY
| | - Alicia A Livinski
- National Institutes of Health Library, Office of Research Services, OD, NIH, Bethesda, MD
| | - Julie S Mak
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA
| | - Joanne Ngeow
- Genomic Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Nicoleta Voian
- Providence Genetic Risk Clinic, Providence Cancer Institute, Portland, OR
| | - Myra J Wick
- Departments of Obstetrics and Gynecology and Clinical Genomics, Mayo Clinic, Rochester, MN
| | - Helen Hanson
- Peninsula Clinical Genetics, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom; Faculty of Health and Life Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Douglas R Stewart
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | - Marc Tischkowitz
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
24
|
Blackford AL, Canto MI, Goggins M. Concerns Regarding the Utility of High-Risk Pancreatic Cancer Surveillance-Reply. JAMA Oncol 2025; 11:79-80. [PMID: 39602156 DOI: 10.1001/jamaoncol.2024.5522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Affiliation(s)
| | | | - Michael Goggins
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
25
|
Du Q, Zhang M, Gao A, He T, Guo M. Epigenetic silencing ZSCAN23 promotes pancreatic cancer growth by activating Wnt signaling. Cancer Biol Ther 2024; 25:2302924. [PMID: 38226836 PMCID: PMC10793710 DOI: 10.1080/15384047.2024.2302924] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/04/2024] [Indexed: 01/17/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most malignant tumor. Zinc finger and SCAN domain-containing protein 23 (ZSCAN23) is a new member of the SCAN domain family. The expression regulation and biological function remain to be elucidated. In this study, we explored the epigenetic regulation and the function of ZSCAN23 in PDAC. ZSCAN23 was methylated in 60.21% (171/284) of PDAC and its expression was regulated by promoter region methylation. The expression of ZSCAN23 inhibited cell proliferation, colony formation, migration, invasion, and induced apoptosis and G1/S phase arrest. ZSCAN23 suppressed Panc10.05 cell xenograft growth in mice. Mechanistically, ZSCAN23 inhibited Wnt signaling by interacting with myosin heavy chain 9 (MYH9) in pancreatic cancer cells. ZSCAN23 is frequently methylated in PDAC and may serve as a detective marker. ZSCAN23 suppresses PDAC cell growth both in vitro and in vivo.
Collapse
Affiliation(s)
- Qian Du
- Department of Gastroenterology and Hepatology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Meiying Zhang
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Aiai Gao
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Tao He
- Department of Pathology, Characteristic Medical Center of the Chinese People's Armed Police Force, Tianjin, People's Republic of China
| | - Mingzhou Guo
- Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- National Key Laboratory of Kidney Diseases, the First Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
26
|
Zhang W, Chen J, Zhang W, Xu M. Advances in Endoscopic Ultrasound in Pancreatic Cancer Screening, Diagnosis, and Palliative Care. Biomedicines 2024; 13:76. [PMID: 39857661 PMCID: PMC11762820 DOI: 10.3390/biomedicines13010076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy with a profoundly poor prognosis. Clinically, the condition most frequently manifests with symptoms including painless jaundice, abdominal discomfort, and back pain. Early diagnosis and the implementation of effective therapeutic strategies are critical for improving patient survival outcomes. However, merely 10-20% of patients are diagnosed at an early stage, with the majority presenting at advanced stages, often with metastasis. Consequently, early detection and intervention are crucial for enhancing prognosis. The widespread adoption of endoscopic ultrasonography (EUS) technology in recent years has significantly enhanced the diagnostic accuracy for pancreatic space-occupying lesions. EUS is increasingly recognized for its pivotal role in alleviating malignant biliary obstruction (MBO), gastric outlet obstruction (GOO), and refractory pain in advanced pancreatic cancer. This article aims to provide an overall review of the current applications of EUS in the diagnosis and treatment of pancreatic cancer, exploring its advantages and limitations in early screening, diagnosis, and palliative care. Furthermore, this review explores potential future directions in the field, aiming to provide valuable insights to inform and enhance the clinical management of pancreatic cancer.
Collapse
Affiliation(s)
- Wenyu Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Jingzheng Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Wei Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- Department of Gastroenterology, Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- Department of Gastroenterology, Digestive Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| |
Collapse
|
27
|
Seyithanoglu D, Durak G, Keles E, Medetalibeyoglu A, Hong Z, Zhang Z, Taktak YB, Cebeci T, Tiwari P, Velichko YS, Yazici C, Tirkes T, Miller FH, Keswani RN, Spampinato C, Wallace MB, Bagci U. Advances for Managing Pancreatic Cystic Lesions: Integrating Imaging and AI Innovations. Cancers (Basel) 2024; 16:4268. [PMID: 39766167 PMCID: PMC11674829 DOI: 10.3390/cancers16244268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/08/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Pancreatic cystic lesions (PCLs) represent a spectrum of non-neoplasms and neoplasms with varying malignant potential, posing significant challenges in diagnosis and management. While some PCLs are precursors to pancreatic cancer, others remain benign, necessitating accurate differentiation for optimal patient care. Conventional approaches to PCL management rely heavily on radiographic imaging, and endoscopic ultrasound (EUS) guided fine-needle aspiration (FNA), coupled with clinical and biochemical data. However, the observer-dependent nature of image interpretation and the complex morphology of PCLs can lead to diagnostic uncertainty and variability in patient management strategies. This review critically evaluates current PCL diagnosis and surveillance practices, showing features of the different lesions and highlighting the potential limitations of conventional methods. We then explore the potential of artificial intelligence (AI) to transform PCL management. AI-driven strategies, including deep learning algorithms for automated pancreas and lesion segmentation, and radiomics for analyzing heterogeneity, can improve diagnostic accuracy and risk stratification. These advanced techniques can provide more objective and reproducible assessments, aiding clinicians in decision-making regarding follow-up intervals and surgical interventions. Early results suggest that AI-driven methods can significantly improve patient outcomes by enabling earlier detection of high-risk lesions and reducing unnecessary procedures for benign cysts. Finally, this review emphasizes that AI-driven approaches could potentially reshape the landscape of PCL management, ultimately leading to improved pancreatic cancer prevention.
Collapse
Affiliation(s)
- Deniz Seyithanoglu
- Machine and Hybrid Intelligence Lab, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (D.S.); (G.D.); (E.K.); (A.M.); (Z.H.); (Z.Z.); (Y.S.V.); (F.H.M.); (R.N.K.)
- Istanbul Faculty of Medicine, Istanbul University, Istanbul 38000, Turkey; (Y.B.T.); (T.C.)
| | - Gorkem Durak
- Machine and Hybrid Intelligence Lab, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (D.S.); (G.D.); (E.K.); (A.M.); (Z.H.); (Z.Z.); (Y.S.V.); (F.H.M.); (R.N.K.)
| | - Elif Keles
- Machine and Hybrid Intelligence Lab, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (D.S.); (G.D.); (E.K.); (A.M.); (Z.H.); (Z.Z.); (Y.S.V.); (F.H.M.); (R.N.K.)
| | - Alpay Medetalibeyoglu
- Machine and Hybrid Intelligence Lab, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (D.S.); (G.D.); (E.K.); (A.M.); (Z.H.); (Z.Z.); (Y.S.V.); (F.H.M.); (R.N.K.)
- Istanbul Faculty of Medicine, Istanbul University, Istanbul 38000, Turkey; (Y.B.T.); (T.C.)
| | - Ziliang Hong
- Machine and Hybrid Intelligence Lab, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (D.S.); (G.D.); (E.K.); (A.M.); (Z.H.); (Z.Z.); (Y.S.V.); (F.H.M.); (R.N.K.)
| | - Zheyuan Zhang
- Machine and Hybrid Intelligence Lab, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (D.S.); (G.D.); (E.K.); (A.M.); (Z.H.); (Z.Z.); (Y.S.V.); (F.H.M.); (R.N.K.)
| | - Yavuz B. Taktak
- Istanbul Faculty of Medicine, Istanbul University, Istanbul 38000, Turkey; (Y.B.T.); (T.C.)
| | - Timurhan Cebeci
- Istanbul Faculty of Medicine, Istanbul University, Istanbul 38000, Turkey; (Y.B.T.); (T.C.)
| | - Pallavi Tiwari
- Department of Radiology, BME, University of Wisconsin-Madison, Madison, WI 53707, USA;
- William S. Middleton Memorial Veterans Affairs (VA) Healthcare, 2500 Overlook Terrace, Madison, WI 53705, USA
| | - Yuri S. Velichko
- Machine and Hybrid Intelligence Lab, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (D.S.); (G.D.); (E.K.); (A.M.); (Z.H.); (Z.Z.); (Y.S.V.); (F.H.M.); (R.N.K.)
| | - Cemal Yazici
- Department of Gastroenterology, University of Illinois at Chicago, Chicago, IL 60611, USA;
| | - Temel Tirkes
- Department of Radiology, Indiana University, Indianapolis, IN 46202, USA;
| | - Frank H. Miller
- Machine and Hybrid Intelligence Lab, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (D.S.); (G.D.); (E.K.); (A.M.); (Z.H.); (Z.Z.); (Y.S.V.); (F.H.M.); (R.N.K.)
| | - Rajesh N. Keswani
- Machine and Hybrid Intelligence Lab, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (D.S.); (G.D.); (E.K.); (A.M.); (Z.H.); (Z.Z.); (Y.S.V.); (F.H.M.); (R.N.K.)
| | - Concetto Spampinato
- Department of Electrical, Electronics and Computer Engineering, University of Catania, 95124 Catania, Italy;
| | - Michael B. Wallace
- Department of Gastroenterology, Mayo Clinic Florida, Jacksonville, FL 32224, USA;
| | - Ulas Bagci
- Machine and Hybrid Intelligence Lab, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (D.S.); (G.D.); (E.K.); (A.M.); (Z.H.); (Z.Z.); (Y.S.V.); (F.H.M.); (R.N.K.)
| |
Collapse
|
28
|
Lyu H, Kong J, Chen J, Zhang R, Xiao S, Guo D, Zhang Q, Chen XZ, Tang J, Zhou C. The Emerging Scenario of Ferroptosis in Pancreatic Cancer Tumorigenesis and Treatment. Int J Mol Sci 2024; 25:13334. [PMID: 39769097 PMCID: PMC11727763 DOI: 10.3390/ijms252413334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 01/05/2025] Open
Abstract
Pancreatic cancer remains one of the most lethal forms of cancer. Currently, there is a lack of effective drug treatments for pancreatic cancer. However, as a newly discovered form of non-apoptotic cell death, ferroptosis has garnered increasing attention in relation to pancreatic cancer. Understanding the role of ferroptosis in the tumorigenesis and treatment of pancreatic cancer may enable more effective clinical trials and treatments for pancreatic cancer and may minimize side effects or restrict the emergence of drug resistance. In this review, we summarize the current knowledge regarding the process and underlying mechanisms of ferroptosis, as well as its dual role in both promoting tumorigenesis and facilitating treatment strategies for pancreatic cancer. Additionally, how ferroptosis is implicated in the development of pancreatitis and insulin resistance, indicating that ferroptosis may play an important role in the risk of pancreatitis- and insulin-resistance-related pancreatic cancers, is also addressed.
Collapse
Affiliation(s)
- Hao Lyu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Jinghua Kong
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Jiasi Chen
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Rui Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Shuai Xiao
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Dong Guo
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Qi Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jingfeng Tang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Cefan Zhou
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| |
Collapse
|
29
|
Harne PS, Harne V, Wray C, Thosani N. Endoscopic innovations in diagnosis and management of pancreatic cancer: a narrative review and future directions. Therap Adv Gastroenterol 2024; 17:17562848241297434. [PMID: 39664230 PMCID: PMC11632891 DOI: 10.1177/17562848241297434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/15/2024] [Indexed: 12/13/2024] Open
Abstract
Pancreatic cancer serves as the third leading cause of cancer-associated morbidity and mortality in the United States, with a 5-year survival rate of only 12% with an expected increase in incidence and mortality in the coming years. Pancreatic ductal adenocarcinomas constitute most pancreatic malignancies. Certain genetic syndromes, including Lynch syndrome, hereditary breast and ovarian cancer syndrome, hereditary pancreatitis, familial adenomatous polyposis, Peutz-Jeghers syndrome, familial pancreatic cancer mutation, and ataxia telangiectasia, confer a significantly higher risk. Screening for pancreatic malignancies currently targets patients with germline mutations or those with significant family history. Screening the general population is not currently viable owing to overall low incidence and lack of specific tests. Endoscopic ultrasound (EUS) and its applied advances are increasingly being used for surveillance, diagnosis, and management of pancreatic malignancies and have now become an indispensable tool in their management. For patients with risk factors, EUS in combination with magnetic resonance imaging/magnetic resonance cholangiopancreatography is used for screening. The role of endoscopic modalities has been expanding with the increased utilization of endoscopic retrograde cholangiopancreatography, EUS-directed therapies include EUS-guided fine-needle aspiration and EUS-fine-needle biopsy (FNB). EUS combined with FNB has the highest specificity and sensitivity for detecting pancreatic cancer amongst available modalities. Studies also recognize that artificial intelligence assisted EUS in the early detection of pancreatic cancer. At the same time, surgical resection has been historically considered the only curative treatment for pancreatic cancer, over 80% of patients present with unresectable disease. We also discuss EUS-guided therapies of physicochemicals (radiofrequency ablation, brachytherapy, and intratumor chemotherapy), biological agents (gene therapies and oncolytic viruses), and immunotherapy. We aim to perform a detailed review of the current burden, risk factors, role of screening, diagnosis, and endoscopic advances in the treatment modalities available for pancreatic cancer.
Collapse
Affiliation(s)
- Prateek Suresh Harne
- Division of Gastroenterology, Allegheny Health Network, Pittsburgh, PA 15212, USA
| | - Vaishali Harne
- Division of Pediatric Gastroenterology, The University of Texas
- Health Science Center and McGovern School of Medicine, Houston, TX, USA
| | - Curtis Wray
- Department of Surgery, The University of Texas Health Science Center and McGovern School of Medicine, Houston, TX, USA
| | - Nirav Thosani
- Department of Surgery and Interventional Gastroenterology, The University of Texas
- Health Science Center and McGovern School of Medicine, Houston, TX, USA
| |
Collapse
|
30
|
Meziani J, de Jong JGY, Fuhler GM, Koopmann BDM, Levink IJM, Fockens P, Vleggaar FP, Bruno MJ, Cahen DL. Assessment of Glucose and HbA1c Monitoring in a Pancreatic Cancer Surveillance Program for High-Risk Individuals. Clin Transl Gastroenterol 2024; 15:e00777. [PMID: 39413349 DOI: 10.14309/ctg.0000000000000777] [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: 04/11/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
INTRODUCTION Several studies suggest that new-onset diabetes mellitus is an early manifestation of pancreatic ductal adenocarcinoma (PDAC). Therefore, the International Cancer of the Pancreas Screening Consortium recommends glucose and hemoglobin A1c (HbA1c) monitoring in high-risk individuals (HRIs) undergoing surveillance. However, evidence that such monitoring improves PDAC detection is lacking. Our aim was to investigate the association between serum glucose and HbA1c values and the development of PDAC in HRIs undergoing surveillance. METHODS Participants were recruited from the familial pancreatic cancer surveillance cohort, which follows hereditary predisposed HRIs yearly by magnetic resonance imaging and/or endoscopic ultrasound and blood sampling. Those who underwent fasting glucose and/or HbA1c monitoring at least once were eligible candidates. RESULTS Four hundred four HRIs met the inclusion criteria. During a median follow-up of 41 months (range 14-120), 9 individuals developed PDAC and 4 (without PDAC) were diagnosed with new-onset diabetes mellitus. Glucose levels ranged from 3.4 to 10.7 mmol/L (mean 5.6 ± 0.7) and HbA1c levels from 25 to 68 mmol/mol (mean 37.7 ± 4.1). The mean values did not differ significantly between PDAC cases and controls. The percentage of individuals with at least one elevated value were comparable between PDAC cases and controls for glucose (33% and 27%, P = 0.707) and HbA1c (22% and 14%, P = 0.623). No consistent glucose or HbA1c trends over time suggested a correlation with PDAC development. DISCUSSION In this HRI surveillance cohort, measuring glucose and HbA1c values did not contribute to PDAC detection. Larger and longer-term studies are needed to determine the final role of glucose and HbA1c monitoring in PDAC surveillance.
Collapse
Affiliation(s)
- Jihane Meziani
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jedidja G Y de Jong
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Gwenny M Fuhler
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Brechtje D M Koopmann
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Iris J M Levink
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Paul Fockens
- Department of Gastroenterology & Hepatology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Frank P Vleggaar
- Department of Gastroenterology & Hepatology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marco J Bruno
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Djuna L Cahen
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
31
|
Xu R, Balmer L, Chen G, Song M. Role of N-Glycosylation in Gastrointestinal Cancers. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:596-607. [PMID: 39514331 DOI: 10.1089/omi.2024.0174] [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: 11/16/2024]
Abstract
Gastrointestinal cancers pose a significant global health challenge. N-glycosylation modulates various cellular processes, including key cancer-related mechanisms. Elucidating its involvement in the onset and advancement of these cancers can offer critical insights for enhancing diagnostic and therapeutic approaches. This review outlines the core process of protein N-glycosylation and highlights its contribution to the progression of gastrointestinal cancers, encompassing cell proliferation, survival, invasion, metastasis, and immune evasion, mainly through its impact on critical signaling pathways. Notably, aberrant N-glycosylation patterns have emerged as crucial biomarkers for the diagnosis and prognosis of various gastrointestinal cancers, providing the foundation for more personalized therapeutic approaches. Therapeutic strategies targeting N-glycosylation, such as glycosyltransferase inhibitors and glycoengineering, show significant promise in mitigating tumor aggressiveness and enhancing immune recognition. However, the clinical implementation of N-glycosylation biomarkers requires the standardization of glycosylation analysis techniques and solutions to challenges in sample processing and data interpretation. Future research efforts should concentrate on overcoming these obstacles to unlock the full potential of N-glycosylation in enhancing cancer management and advancing patient outcomes.
Collapse
Affiliation(s)
- Ruirui Xu
- Center for Precision Health, Edith Cowan University, Western Australia, Australia
- School of Medical and Health Science, Edith Cowan University, Western Australia, Australia
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Shantou University Medical College, Guangdong, China
| | - Lois Balmer
- Center for Precision Health, Edith Cowan University, Western Australia, Australia
- School of Medical and Health Science, Edith Cowan University, Western Australia, Australia
| | - Gengzhen Chen
- Digestive Disease Prevention and Treatment Center, Chenghai District People's Hospital, Guangdong, China
| | - Manshu Song
- School of Medical and Health Science, Edith Cowan University, Western Australia, Australia
| |
Collapse
|
32
|
Millastre J, Hermoso-Durán S, Solórzano MOD, Fraunhoffer N, García-Rayado G, Vega S, Bujanda L, Sostres C, Lanas Á, Velázquez-Campoy A, Abian O. Thermal Liquid Biopsy: A Promising Tool for the Differential Diagnosis of Pancreatic Cystic Lesions and Malignancy Detection. Cancers (Basel) 2024; 16:4024. [PMID: 39682210 DOI: 10.3390/cancers16234024] [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: 10/30/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Pancreatic cystic lesions (PCLs) are a heterogeneous group of lesions with increasing incidence, usually identified incidentally on imaging studies (multidetector computed tomography (MDCT), magnetic resonance imaging (MRI), or endoscopic ultrasound (EUS)) [...].
Collapse
Affiliation(s)
- Judith Millastre
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Sonia Hermoso-Durán
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - María Ortiz de Solórzano
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Nicolas Fraunhoffer
- Programa Franco-Argentino de Estudio del Cáncer de Páncreas, Buenos Aires, Argentina
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, CNRS UMR, Aix-Marseille Université, 13009 Marseille, France
- Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Equipe Labellisée La Ligue, 13288 Marseille, France
| | - Guillermo García-Rayado
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Sonia Vega
- Institute for Biocomputation and Physics of Complex Systems, University of Zaragoza, 50018 Zaragoza, Spain
| | - Luis Bujanda
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
- Donostia University Hospital, University of the Basque Country (UPV/EHU), 20014 San Sebastian, Spain
| | - Carlos Sostres
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Ángel Lanas
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, 50009 Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Adrián Velázquez-Campoy
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
- Institute for Biocomputation and Physics of Complex Systems, University of Zaragoza, 50018 Zaragoza, Spain
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, 50009 Zaragoza, Spain
| | - Olga Abian
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
- Institute for Biocomputation and Physics of Complex Systems, University of Zaragoza, 50018 Zaragoza, Spain
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, 50009 Zaragoza, Spain
| |
Collapse
|
33
|
Llach J, Luzko I, Earl J, Barreto E, Rodríguez-Garrote M, Lleixà M, Herrera-Pariente C, Fernández G, Munoz J, Bonjoch L, Saurí T, Ausania F, Ocaña T, Moreno L, Grau E, Oriola J, Alvarez-Mora MI, Herreros-Villanueva M, Castellví-Bel S, Balaguer F, Bujanda L, Moreira L. Should We Offer Universal Germline Genetic Testing to All Patients with Pancreatic Cancer? A Multicenter Study. Cancers (Basel) 2024; 16:3779. [PMID: 39594734 PMCID: PMC11592128 DOI: 10.3390/cancers16223779] [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: 09/28/2024] [Revised: 11/01/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is associated with a significant percentage of germline pathogenic variants (GPVs). Unlike in the United States, routine universal genetic testing is not performed in Europe. The aim of the study is to evaluate the diagnostic yield of germline genetic testing in all patients with PDAC. METHODS Individuals with newly diagnosed PDAC from three Spanish hospitals were enrolled, regardless of family history. Thirteen known susceptibility genes for PDAC were studied using a multigene panel or whole-exome sequencing. RESULTS One hundred seventy-nine PDAC patients underwent genetic testing. Fourteen (7.8%) had a GPV or likely pathogenic variant In the genes studied: six in ATM, six in BRCA2, one in PALB2, and one in TP53. Of these, seven (50%) did not meet the clinical criteria for genetic study and would have been classified as sporadic PDAC. Presenting with a personal history of any other neoplasm was associated with some GPV, with an odds ratio (OR) of 3.5 (1.1-11.5). A family history of PDAC and breast cancer was also associated with some GPV, with oRs of 3.7 (1.08-13.6) and 8.5 (2.6-26.6), respectively. None of the patients over 60 years without a relevant family history of malignancies presented a GPV associated with PDAC. CONCLUSIONS In our PDAC cohort, a noteworthy number of GPVs were identified, and half of these patients would have been classified as sporadic based solely on clinical criteria. Genetic testing should always be considered, particularly in patients under 60 years or those with a history of other malignancies, especially where economic resources need optimization.
Collapse
Affiliation(s)
- Joan Llach
- Department of Gastroenterology, Hospital Clínic Barcelona, 08036 Barcelona, Spain; (J.L.); (I.L.); (M.L.); (J.M.); (T.O.); (L.M.); (E.G.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain; (C.H.-P.); (L.B.); (S.C.-B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
- Campus Clínic, University of Barcelona, 08036 Barcelona, Spain
| | - Irina Luzko
- Department of Gastroenterology, Hospital Clínic Barcelona, 08036 Barcelona, Spain; (J.L.); (I.L.); (M.L.); (J.M.); (T.O.); (L.M.); (E.G.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain; (C.H.-P.); (L.B.); (S.C.-B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
- Campus Clínic, University of Barcelona, 08036 Barcelona, Spain
| | - Julie Earl
- Ramón y Cajal Health Research Institute (IRYCIS), The Biomedical Research Network in Cancer (CIBERONC), 28029 Madrid, Spain; (J.E.); (E.B.); (M.R.-G.)
| | - Emma Barreto
- Ramón y Cajal Health Research Institute (IRYCIS), The Biomedical Research Network in Cancer (CIBERONC), 28029 Madrid, Spain; (J.E.); (E.B.); (M.R.-G.)
- School of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28805 Alcalá de Henares, Spain
| | - Mercedes Rodríguez-Garrote
- Ramón y Cajal Health Research Institute (IRYCIS), The Biomedical Research Network in Cancer (CIBERONC), 28029 Madrid, Spain; (J.E.); (E.B.); (M.R.-G.)
- School of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28805 Alcalá de Henares, Spain
| | - Marc Lleixà
- Department of Gastroenterology, Hospital Clínic Barcelona, 08036 Barcelona, Spain; (J.L.); (I.L.); (M.L.); (J.M.); (T.O.); (L.M.); (E.G.); (F.B.)
| | - Cristina Herrera-Pariente
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain; (C.H.-P.); (L.B.); (S.C.-B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
| | - Guerau Fernández
- Department of Genetic and Molecular Medicine-IPER, Hospital Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Center for Biomedical Research Network on Rare Diseases (CIBERER), 08036 Barcelona, Spain;
| | - Jenifer Munoz
- Department of Gastroenterology, Hospital Clínic Barcelona, 08036 Barcelona, Spain; (J.L.); (I.L.); (M.L.); (J.M.); (T.O.); (L.M.); (E.G.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain; (C.H.-P.); (L.B.); (S.C.-B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
| | - Laia Bonjoch
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain; (C.H.-P.); (L.B.); (S.C.-B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
| | - Tamara Saurí
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
- Campus Clínic, University of Barcelona, 08036 Barcelona, Spain
- Medical Oncology Department, Hospital Clinic and Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, 08036 Barcelona, Spain
| | - Fabio Ausania
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
- Campus Clínic, University of Barcelona, 08036 Barcelona, Spain
- Department of General and Digestive Surgery, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| | - Teresa Ocaña
- Department of Gastroenterology, Hospital Clínic Barcelona, 08036 Barcelona, Spain; (J.L.); (I.L.); (M.L.); (J.M.); (T.O.); (L.M.); (E.G.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain; (C.H.-P.); (L.B.); (S.C.-B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
| | - Lorena Moreno
- Department of Gastroenterology, Hospital Clínic Barcelona, 08036 Barcelona, Spain; (J.L.); (I.L.); (M.L.); (J.M.); (T.O.); (L.M.); (E.G.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain; (C.H.-P.); (L.B.); (S.C.-B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
| | - Elia Grau
- Department of Gastroenterology, Hospital Clínic Barcelona, 08036 Barcelona, Spain; (J.L.); (I.L.); (M.L.); (J.M.); (T.O.); (L.M.); (E.G.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain; (C.H.-P.); (L.B.); (S.C.-B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
| | - Josep Oriola
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
- Biochemistry and Molecular Genetics Department, CDB, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Maria Isabel Alvarez-Mora
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
- Biochemistry and Molecular Genetics Department, CDB, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Marta Herreros-Villanueva
- Facultad de Ciencias de la Salud, Universidad Isabel I, 09003 Burgos, Spain;
- Department of Gastroenterology, Hospital Donostia, 20014 San Sebastián, Spain
- Instituto Biodonostia, 20014 San Sebastián, Spain
| | - Sergi Castellví-Bel
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain; (C.H.-P.); (L.B.); (S.C.-B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
| | - Francesc Balaguer
- Department of Gastroenterology, Hospital Clínic Barcelona, 08036 Barcelona, Spain; (J.L.); (I.L.); (M.L.); (J.M.); (T.O.); (L.M.); (E.G.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain; (C.H.-P.); (L.B.); (S.C.-B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
- Campus Clínic, University of Barcelona, 08036 Barcelona, Spain
| | - Luis Bujanda
- Department of Gastroenterology, Biogipuzkoa Health Research Institute, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco (UPV/EHU), 20014 San Sebastián, Spain;
| | - Leticia Moreira
- Department of Gastroenterology, Hospital Clínic Barcelona, 08036 Barcelona, Spain; (J.L.); (I.L.); (M.L.); (J.M.); (T.O.); (L.M.); (E.G.); (F.B.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 08036 Barcelona, Spain; (C.H.-P.); (L.B.); (S.C.-B.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (T.S.); (F.A.); (J.O.); (M.I.A.-M.)
- Campus Clínic, University of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
34
|
Pandharipande PV, Kang SK. How Do We Assess Controversies Using Evidence-Based Radiology? Radiol Clin North Am 2024; 62:929-934. [PMID: 39393851 DOI: 10.1016/j.rcl.2024.05.002] [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: 10/13/2024]
Abstract
At its best, the practice of medicine involves careful integration of experience and evidence. Generating evidence to address controversies in radiology - and translating such evidence to practice - requires appropriate selection of methods, and an understanding of the strengths, shortcomings, and biases inherent to different research designs and analyses. Equipped with such knowledge, the radiologic community can ensure that both research and clinical practice in our discipline excels, and that those questions that will be the most critical to answer will be formulated for successful investigation in the years to come.
Collapse
Affiliation(s)
- Pari V Pandharipande
- The Ohio State University College of Medicine, 450 Faculty Tower, 395 West 12th Street, 4th Floor, Columbus, OH 43210, USA.
| | - Stella K Kang
- Population Health Imaging and Outcomes, NYU Department of Radiology, 660 First Avenue, Room 333, New York, NY 10016, USA
| |
Collapse
|
35
|
Doi T, Ishikawa T, Sakakida T, Itani J, Sone D, Morita R, Kataoka S, Miyake H, Seko Y, Yamaguchi K, Moriguchi M, Sogame Y, Konishi H, Murashima K, Iwasaku M, Takayama K, Itoh Y. Real-world genome profiling in Japanese patients with pancreatic ductal adenocarcinoma focusing on HRD implications. Cancer Sci 2024; 115:3729-3739. [PMID: 39315592 PMCID: PMC11531956 DOI: 10.1111/cas.16329] [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: 06/09/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/25/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses significant challenges due to its high mortality, making it a critical area of research. This retrospective observational study aimed to analyze real-world data from comprehensive genome profiling (CGP) of Japanese patients with PDAC, mainly focusing on differences in gene detection rates among panels and the implications for homologous recombination deficiency (HRD) status. This study enrolled 2568 patients with PDAC who had undergone CGP between June 2019 and December 2021 using data from the nationwide Center for Cancer Genomics and Advanced Therapeutics database. Two types of CGP assays (tissue and liquid biopsies) were compared and a higher detection rate of genetic abnormalities in tissue specimens was revealed. HRD-related gene alterations were detected in 23% of patients, with BRCA1/2 mutations accounting for 0.9% and 2.9% of patients, respectively. Treatment outcome analysis indicated that patients with BRCA1/2 mutations had a longer time to treatment discontinuation with FOLFIRINOX than gemcitabine plus nab-paclitaxel as first-line therapy (9.3 vs. 5.6 months, p = 0.028). However, no significant differences were observed in the treatment response among the other HRD-related genes. Logistic regression analysis identified younger age and family history of breast, prostate, and ovarian cancers as predictive factors for HRD-related gene alterations. Despite the lack of progression-free survival data and the inability to discriminate between germline and somatic mutations, this study provides valuable insights into the clinical implications of CGP in Japanese patients with PDAC. Further research is warranted to optimize panel selection and elucidate the efficacy of platinum-based therapies depending on the HRD status.
Collapse
Affiliation(s)
- Toshifumi Doi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
| | - Takeshi Ishikawa
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
- Department of Medical Oncology UnitUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
| | - Tomoki Sakakida
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
| | - Junichiro Itani
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
| | - Daiki Sone
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
| | - Ryuichi Morita
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
| | - Seita Kataoka
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Hayato Miyake
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Yuya Seko
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Kanji Yamaguchi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Michihisa Moriguchi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Yoshio Sogame
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Hideyuki Konishi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Kyoko Murashima
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
| | - Masahiro Iwasaku
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
- Department of Pulmonary Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Koichi Takayama
- Cancer Genome Medical CenterUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
- Department of Medical Oncology UnitUniversity Hospital, Kyoto Prefectural University of MedicineKyotoJapan
- Department of Pulmonary Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| |
Collapse
|
36
|
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
|
37
|
McDonald HG, Kennedy A, Solomon AL, Williams CM, Reagan AM, Cassim E, Harper M, Burke E, Armstrong T, Gosky M, Cavnar M, Pandalai PK, Barry-Hundeyin M, Patel R, Nutalapati S, Moss J, Hull PC, Kolesar J, Pickarski JC, Kim J. Development of a Novel Protocol for Germline Testing in Pancreatic Cancer. Ann Surg Oncol 2024; 31:7705-7712. [PMID: 39133448 DOI: 10.1245/s10434-024-16011-3] [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: 05/20/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Guidelines now recommend universal germline genetic testing (GGT) for all pancreatic ductal adenocarcinoma (PDAC) patients. Testing provides information on actionable pathogenic variants and guides management of patients and family. Since traditional genetic counseling (GC) models are time-intensive and GC resources are sparse, new approaches are needed to comply with guidelines without overwhelming available resources. METHODS A novel protocol was developed for physician-led GGT. Completed test kits were delivered to the GC team, who maintained a prospective database and mailed all orders. If results revealed pathogenic variants for PDAC, patients were offered comprehensive GC, whereas negative and variant of uncertain significance (VUS) test results were reported to patients via brief calls. RESULTS During protocol implementation between January 2020 and December 2022, 310 (81.5%) patients underwent GGT, with a physician compliance rate of 82.6% and patient compliance rate of 98.7%. Of 310 patients tested, 44 (14.2%) patients had detection of pathogenic variants, while 83 (26.8%) patients had VUS. Pathogenic variants included BRCA1/BRCA2/PALB2 (n = 18, 5.8%), ATM (n = 9, 2.9%), CFTR (n = 4, 1.3%), EPCAM/MLH1/MSH2/MSH6/PMS2 (n = 3, 1.0%), and CDKN2A (n = 2, 0.7%). The GC team successfully contacted all patients with pathogenic variants to discuss results and offer comprehensive GC. CONCLUSION Our novel protocol facilitated GGT with excellent compliance despite limited GC resources. This framework for GGT allocates GC resources to those patients who would benefit most from GC. As we continue to expand the program, we seek to implement methods to ensure compliance with cascade testing of high-risk family members.
Collapse
Affiliation(s)
- Hannah G McDonald
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Andrew Kennedy
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Angelica L Solomon
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Chelsey M Williams
- Division of Hematology Oncology, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Anna M Reagan
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Emily Cassim
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Megan Harper
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Erin Burke
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Terra Armstrong
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Michael Gosky
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Michael Cavnar
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Prakash K Pandalai
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Mautin Barry-Hundeyin
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Reema Patel
- Division of Hematology Oncology, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Snigdha Nutalapati
- Division of Hematology Oncology, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Jessica Moss
- Division of Hematology Oncology, Department of Medicine, University of Kentucky, Lexington, KY, USA
| | - Pamela C Hull
- Department of Behavioral Science, University of Kentucky, Lexington, KY, USA
| | - Jill Kolesar
- College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | | | - Joseph Kim
- Division of Surgical Oncology, Department of Surgery, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
38
|
Rodríguez-Olivares JL, Kimball TN, Jeter JM, De-La-Mora-Molina H, Núñez I, Weitzel JN, Chávarri-Guerra Y. Prevalence and spectrum of germline pathogenic variants in cancer susceptibility genes among mexican patients with exocrine pancreatic cancer. Pancreatology 2024; 24:1049-1056. [PMID: 39327123 DOI: 10.1016/j.pan.2024.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Although universal germline genetic testing is recommended for patients with exocrine pancreatic cancer (PC), access to genetic testing remains limited in low- and middle-income countries. This study aims to narrow the gap in our understanding of the spectrum of germline pathogenic and likely pathogenic variants (PVs) in cancer susceptibility genes in the Mexican population. METHODS The landscape of PVs in cancer susceptibility genes was identified by next-generation sequencing multigene panel assays among patients with PC who were enrolled in the Clinical Cancer Genomics Community Research Network prospective registry in Mexico City. RESULTS From August 2019 to April 2023, 137 patients underwent genetic testing. The median age at diagnosis was 60 years (range 36-85), 58.4 % were women, and 38.7 % were metastatic at diagnosis. The frequency of germline PVs was 16 % (n = 22): ATM 36.4 % (n = 8), CDKN2A/p16INK4A 27.3 % (n = 6), BRCA2 9.1 % (n = 2), PALB2 9.1 % (n = 2), CHEK2 9.1 % (n = 2), TP53 4.5 % (n = 1), and NF1 4.5 % (n = 1). Additionally, 2 carriers of monoallelic germline variants in MUTYH were identified. No significant differences were observed between carriers and non-carriers in terms of family history of pancreatic cancer. CONCLUSIONS We identified a significant frequency of actionable germline PVs in Mexicans with PC, wherein the majority were in a broad spectrum of genes associated with the homologous recombination DNA repair mechanism. Most pancreatic cancer associated PVs were detected in non-BRCA genes, so our findings support the recommendation of multigene panel testing for genetic cancer risk assessment of Mexican individuals with PC.
Collapse
Affiliation(s)
- José Luis Rodríguez-Olivares
- Department of Hematology and Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Tamara N Kimball
- Center for Genomic Medicine. Massachusetts General Hospital, Boston, MA, USA
| | - Joanne M Jeter
- Department of Oncology. City of Hope Cancer Center, Duarte, CA, USA
| | - Héctor De-La-Mora-Molina
- Department of Hematology and Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Isaac Núñez
- Research Division. Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jeffrey N Weitzel
- Division of Precision Prevention, University of Kansas Comprehensive Cancer Center, Kansas City, USA
| | - Yanin Chávarri-Guerra
- Department of Hematology and Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
39
|
M'Baloula J, Tougeron D, Boilève A, Jeanbert E, Guimbaud R, Ben Abdelghani M, Durand A, Turpin A, Quesada S, Blanc JF, Artru P, Toullec C, Trouilloud I, Pellat A, Touchefeu Y, Pinot J, Caroli-Bosc FX, Taïeb J, Doat S, Bouché O, Védie AL, de Mestier L, Muller M. Olaparib as maintenance therapy in non resectable pancreatic adenocarcinoma associated with homologous recombination deficiency profile: A French retrospective multicentric AGEO real-world study. Eur J Cancer 2024; 212:115051. [PMID: 39366210 DOI: 10.1016/j.ejca.2024.115051] [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: 07/19/2024] [Revised: 09/13/2024] [Accepted: 09/22/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis. The POLO trial showed that olaparib (PARP inhibitor) improved progression-free survival (PFS) but not overall survival (OS), when used as maintenance therapy after ≥ 16 weeks of disease control with first-line platinum-based chemotherapy in patients with germline (g) BRCA 1 or 2 pathogenic variants (PV) metastatic PDAC. However, real-world data on the effectiveness of olaparib are missing. METHODS Patients with unresectable PDAC associated with somatic (s) or (g)BRCA1/2 and (g)non-BRCA-HRD PV (i.e. other homologous recombination deficiency/HRD genes) who were treated with olaparib between 2020-2023 were included. The primary objective was to describe treatment patterns. Secondary exploratory objectives included OS and PFS in patients treated with olaparib according to the POLO trial or not, OS and PFS in patients with (g)HRD PV-associated PDAC versus (s)PVs, olaparib safety profile and factors associated with olaparib poor outcomes. RESULTS Among 85 patients, 45.9 % received olaparib as defined by the POLO trial. No difference in OS and PFS was observed between patients who received olaparib according to the POLO trial versus not. Patients with (g)HRD PV-associated PDAC had better OS compared to others (22.3 versus 10.5 months, p = 0.038). Factors associated with olaparib poor outcomes included a high neutrophil-to-lymphocyte ratio and the use of olaparib outside the recommendations of the POLO trial. Few grade ≥ 3 adverse events were reported (9.4 %). CONCLUSION Patients with (g)HRD PV-associated PDAC had longer OS than those with (s)HRD PV. Olaparib use beyond the scope of the POLO trial was associated with poor outcomes.
Collapse
Affiliation(s)
- Jeannie M'Baloula
- University of Lorraine, Department of Hepato-Gastroenterology and digestive oncology, Nancy University Hospital, Nancy, France
| | - David Tougeron
- Department of Hepatology and Gastroenterology, Poitiers University Hospital, Poitiers, France
| | - Alice Boilève
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Elodie Jeanbert
- DRCI, MPI Department, Methodology, Data Management and Statistics Unit, Nancy University Hospital, Nancy, France
| | - Rosine Guimbaud
- Digestive Medical Oncology, Toulouse University Hospital, Toulouse, France
| | | | - Alice Durand
- Department of Gastroenterology and digestive oncology, Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon, France
| | - Anthony Turpin
- Medical Oncology Department, Lille University Hospital, University of Lille, Lille, France
| | - Stanislas Quesada
- Medical Oncology Department, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Jean Frédéric Blanc
- Department of Gastroenterology and digestive oncology, Hôpital Haut-Lévêque, Bordeaux University Hospital, Bordeaux, France
| | - Pascal Artru
- kHepato-gastroenterology department, Hôpital Jean-Mermoz, Lyon, France
| | - Clémence Toullec
- Medical Oncology Department, Sainte Catherine Institut du Cancer Avignon-Provence, Avignon, France
| | - Isabelle Trouilloud
- Department of Medical Oncology, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, Sorbonne University, Paris, France
| | - Anna Pellat
- Department of Gastroenterology, endoscopy and digestive oncology, Assistance Publique-Hôpitaux de Paris, Cochin University Hospital, Paris, France
| | - Yann Touchefeu
- Hepato-gastroenterology Department, Nantes University Hospital, Nantes, Institut des Maladies de l'Appareil Digestif (IMAD), Inserm CIC, 1413 Nantes, France
| | - Julien Pinot
- Department of Medical Oncology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | | | - Julien Taïeb
- Department of Gastroenterology and digestive oncology, Assistance Publique-Hôpitaux de Paris, Georges Pompidou European Hospital, CARPEM comprehensive cancer center, Université Paris Cité, Paris, France
| | - Solène Doat
- Department of Gastroenterology, Assistance Publique-Hôpitaux de Paris, Pitié-Salpetrière Hospital, Paris, France
| | - Olivier Bouché
- Department of Hepato-Gastroenterology and digestive oncology, Robert-Debré University Hospital, Reims, France
| | - Anne Laure Védie
- University Paris-Cité, Department of Pancreatology and digestive oncology, Beaujon Hospital (AP-HP), Clichy, France
| | - Louis de Mestier
- University Paris-Cité, Department of Pancreatology and digestive oncology, Beaujon Hospital (AP-HP), Clichy, France
| | - Marie Muller
- University of Lorraine, Department of Hepato-Gastroenterology and digestive oncology, Nancy University Hospital, Nancy, France.
| |
Collapse
|
40
|
García García de Paredes A, Martínez Moneo E, Lariño-Noia J, Earl J. Pancreatic cancer screening in high-risk individuals. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2024; 116:519-522. [PMID: 39087662 DOI: 10.17235/reed.2024.10635/2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
The incidence of pancreatic cancer is increasing, although globally it represents less than 3% of all cancers. Despite advances in medical and surgical management, survival rates have not significantly improved in recent years. Consequently, pancreatic cancer, though relatively uncommon, is the third leading cause of cancer-related deaths. This is primarily due to the disease´s late detection. Symptoms appear late and are nonspecific, and over 80% of cases are diagnosed at an advanced stage and unsuitable for curative surgery, resulting in a five-year survival rate below 10%. However, the exceptional cases that are diagnosed early show five-year survival rates exceeding 80%. Therefore, one of the keys to improving pancreatic cancer prognosis lies in early detection, making screening in high-risk individuals a potentially crucial strategy.
Collapse
Affiliation(s)
| | | | | | - Julie Earl
- Biomarkers and Personalized Approach to Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)
| |
Collapse
|
41
|
Yasinzai AQK, Tareen B, Tracy K, Jamil N, Khan M, Ullah H, Raza M, Khan AU, Arif D, Waheed A, Sidhwa F, Misra S, Karki NR, Karim NA, Cavalcante L, Ullah A. Pancreatic ductal adenocarcinoma: exploring clinicopathological trends and racial disparities in a comprehensive U.S. population-based study. Clin Transl Oncol 2024; 26:2618-2628. [PMID: 38615292 DOI: 10.1007/s12094-024-03484-7] [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: 01/25/2024] [Accepted: 03/25/2024] [Indexed: 04/15/2024]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy about 50% of PDAC are metastatic at presentation. In this study, we evaluated PDAC demographics, annual trend analysis, racial disparities, survival rate, and the role of different treatment modalities in localized and metastatic disease. METHODS A total of 144,824 cases of PDAC were obtained from the SEER database from 2000 to 2018. RESULTS The median age was 69 years, with a slightly higher incidence in males (52%) and 80% of all cases were white. Among cases with available data, 43% were grade III tumors and 57% were metastatic. The most common site of metastasis was the liver (15.7%). The annual incidence has increased steadily from 2000 to 2018. The overall observed (OS) 5-year survival rate was 4.4% (95% CI 4.3-4.6%), and 5 years cause-specific survival (CSS) was 5% (95% CI 5.1-5.4%). The 5-year survival with multimodal therapy (chemotherapy, surgery, and radiation) was 22% (95% CI 20.5-22.8%). 5-year CSS for the blacks was lower at 4.7% (95% CI 4.2-5.1%) compared to the whites at 5.3% (95% CI 5.1-5.4%). Multivariate analysis found male gender and black race associated with worse prognosis. Kaplan-Meier survival analysis found multimodal therapy to have the best outcomes in all three stages. CONCLUSION PDAC is an aggressive malignancy with male gender and black race are associated with a poor prognosis. Surgery with chemoradiation was associated with the best overall survival. With steadily increasing rates of PDAC, improved treatment modalities are paramount to improving survival in these patients.
Collapse
Affiliation(s)
| | - Bisma Tareen
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | | | - Nimra Jamil
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Marjan Khan
- Internal Medicine, Marshfield Medical Center, Marshfield, USA
| | - Hafeez Ullah
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Muhammad Raza
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Amin Ullah Khan
- Department of Medicine, Bolan Medical College, Quetta, 83700, Pakistan
| | - Dauod Arif
- Department of Oncology Developmental Therapeutics, Novant Health, Charlotte, NC, USA
| | - Abdul Waheed
- Department of Surgery, San Joaquin General Hospital, French Camp, CA, 95231, USA
| | - Feroze Sidhwa
- Department of Surgery, San Joaquin General Hospital, French Camp, CA, 95231, USA
| | - Subhasis Misra
- Department of Surgery, BayCare Health System, Tampa, FL, USA
| | - Nabin Raj Karki
- Department of Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, USA
| | - Nagla Abdel Karim
- Department of Hematology-Oncology, Inova Schar Cancer Institute, University of Virginia, Fairfax, VA, 22031, USA
| | - Ludimila Cavalcante
- Department of Oncology Developmental Therapeutics, Novant Health, Charlotte, NC, USA
| | - Asad Ullah
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| |
Collapse
|
42
|
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
|
43
|
Kawai M, Fukuda A, Otomo R, Obata S, Minaga K, Asada M, Umemura A, Uenoyama Y, Hieda N, Morita T, Minami R, Marui S, Yamauchi Y, Nakai Y, Takada Y, Ikuta K, Yoshioka T, Mizukoshi K, Iwane K, Yamakawa G, Namikawa M, Sono M, Nagao M, Maruno T, Nakanishi Y, Hirai M, Kanda N, Shio S, Itani T, Fujii S, Kimura T, Matsumura K, Ohana M, Yazumi S, Kawanami C, Yamashita Y, Marusawa H, Watanabe T, Ito Y, Kudo M, Seno H. Early detection of pancreatic cancer by comprehensive serum miRNA sequencing with automated machine learning. Br J Cancer 2024; 131:1158-1168. [PMID: 39198617 PMCID: PMC11442445 DOI: 10.1038/s41416-024-02794-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 06/26/2024] [Accepted: 07/03/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Pancreatic cancer is often diagnosed at advanced stages, and early-stage diagnosis of pancreatic cancer is difficult because of nonspecific symptoms and lack of available biomarkers. METHODS We performed comprehensive serum miRNA sequencing of 212 pancreatic cancer patient samples from 14 hospitals and 213 non-cancerous healthy control samples. We randomly classified the pancreatic cancer and control samples into two cohorts: a training cohort (N = 185) and a validation cohort (N = 240). We created ensemble models that combined automated machine learning with 100 highly expressed miRNAs and their combination with CA19-9 and validated the performance of the models in the independent validation cohort. RESULTS The diagnostic model with the combination of the 100 highly expressed miRNAs and CA19-9 could discriminate pancreatic cancer from non-cancer healthy control with high accuracy (area under the curve (AUC), 0.99; sensitivity, 90%; specificity, 98%). We validated high diagnostic accuracy in an independent asymptomatic early-stage (stage 0-I) pancreatic cancer cohort (AUC:0.97; sensitivity, 67%; specificity, 98%). CONCLUSIONS We demonstrate that the 100 highly expressed miRNAs and their combination with CA19-9 could be biomarkers for the specific and early detection of pancreatic cancer.
Collapse
Affiliation(s)
- Munenori Kawai
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan.
| | - Ryo Otomo
- Research and Development Division, ARKRAY, Inc., Yousuien-nai, 59 Gansuin-cho, Kamigyo-ku, Kyoto, Japan
| | - Shunsuke Obata
- Research and Development Division, ARKRAY, Inc., Yousuien-nai, 59 Gansuin-cho, Kamigyo-ku, Kyoto, Japan
| | - Kosuke Minaga
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Masanori Asada
- Department of Gastroenterology and Hepatology, Osaka Red Cross Hospital, Osaka, Japan
| | - Atsushi Umemura
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Uenoyama
- Department of Gastroenterology and Hepatology, Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
| | - Nobuhiro Hieda
- Department of Gastroenterology, Otsu Red Cross Hospital, Shiga, Japan
| | - Toshihiro Morita
- Department of Gastroenterology and Hepatology, Kitano Hospital, Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Ryuki Minami
- Department of Gastroenterology, Tenri Hospital, Nara, Japan
| | - Saiko Marui
- Department of Gastroenterology and Hepatology, Shiga General Hospital, Shiga, Japan
| | - Yuki Yamauchi
- Department of Gastroenterology, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Japan
| | - Yoshitaka Nakai
- Department of Gastroenterology and Hepatology, Kyoto Katsura Hospital, Kyoto, Japan
| | - Yutaka Takada
- Department of Gastroenterology and Hepatology, Kobe City Nishi-Kobe Medical Center, Kobe, Japan
| | - Kozo Ikuta
- Division of Gastroenterology, Shinko Hospital, Kobe, Japan
| | - Takuto Yoshioka
- Department of Gastroenterology and Hepatology, Takatsuki Red Cross Hospital, Takatsuki, Japan
| | - Kenta Mizukoshi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Kosuke Iwane
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Go Yamakawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Mio Namikawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Makoto Sono
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Munemasa Nagao
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Takahisa Maruno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Yuki Nakanishi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| | - Mitsuharu Hirai
- Research and Development Division, ARKRAY, Inc., Yousuien-nai, 59 Gansuin-cho, Kamigyo-ku, Kyoto, Japan
| | - Naoki Kanda
- Department of Gastroenterology and Hepatology, Takatsuki Red Cross Hospital, Takatsuki, Japan
| | - Seiji Shio
- Division of Gastroenterology, Shinko Hospital, Kobe, Japan
| | - Toshinao Itani
- Department of Gastroenterology and Hepatology, Kobe City Nishi-Kobe Medical Center, Kobe, Japan
| | - Shigehiko Fujii
- Department of Gastroenterology and Hepatology, Kyoto Katsura Hospital, Kyoto, Japan
| | - Toshiyuki Kimura
- Department of Gastroenterology, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Japan
| | - Kazuyoshi Matsumura
- Department of Gastroenterology and Hepatology, Shiga General Hospital, Shiga, Japan
| | - Masaya Ohana
- Department of Gastroenterology, Tenri Hospital, Nara, Japan
| | - Shujiro Yazumi
- Department of Gastroenterology and Hepatology, Kitano Hospital, Tazuke Kofukai Medical Research Institute, Osaka, Japan
| | - Chiharu Kawanami
- Department of Gastroenterology, Otsu Red Cross Hospital, Shiga, Japan
| | - Yukitaka Yamashita
- Department of Gastroenterology and Hepatology, Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
| | - Hiroyuki Marusawa
- Department of Gastroenterology and Hepatology, Osaka Red Cross Hospital, Osaka, Japan
| | - Tomohiro Watanabe
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yoshito Ito
- Department of Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
44
|
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; 62:1724-1785. [PMID: 39389105 DOI: 10.1055/a-2338-3716] [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
|
45
|
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
|
46
|
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:e874-e995. [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
|
47
|
Oppat KM, Bennett FJ, Maithel SK. A Review of the Indications, Outcomes, and Postoperative Management After Total and Completion Pancreatectomy for Pancreatic Cancer: More Is Not Necessarily Better. Surg Clin North Am 2024; 104:1049-1064. [PMID: 39237163 PMCID: PMC11889495 DOI: 10.1016/j.suc.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
With improvements in surgical technique and advances in pancreatic endocrine and exocrine replacement therapy, the indications for, and threshold to perform, total or completion pancreatectomy in the modern surgical era are ever evolving. The following review will evaluate such indications for pancreatic cancer including pancreatic ductal adenocarcinoma and intraductal papillary mucinous neoplasms. The authors also review the literature on oncologic outcomes of total and completion pancreatectomy for pancreatic cancer. Finally, they discuss the quality of life and postoperative management of the a-pancreatic state.
Collapse
Affiliation(s)
- Kailey M Oppat
- Emory University, 1365B Clifton Road, NE Building B, Suite 4100, Office 4202, Atlanta, GA 30302, USA
| | - Frances J Bennett
- Emory University, 1365B Clifton Road, NE Building B, Suite 4100, Office 4202, Atlanta, GA 30302, USA
| | - Shishir K Maithel
- Emory University, 1365B Clifton Road, NE Building B, Suite 4100, Office 4202, Atlanta, GA 30302, USA.
| |
Collapse
|
48
|
Leader AE, Rebbeck TR, Oh WK, Patel AV, Winer EP, Bailey LO, Gomella LG, Lumpkins CY, Garraway IP, Aiello LB, Baskin ML, Cheng HH, Cooney KA, Ganzak A, George DJ, Halabi S, Hathaway F, Healy C, Kim JW, Leapman MS, Loeb S, Maxwell KN, McNair C, Morgan TM, Prindeville B, Soule HR, Steward WL, Suttiratana SC, Taplin ME, Yamoah K, Fortune T, Bennett K, Blanding-Godbolt J, Gross L, Giri VN. Adaptation of the socioecological model to address disparities in engagement of Black men in prostate cancer genetic testing. BMC Public Health 2024; 24:2533. [PMID: 39289635 PMCID: PMC11409532 DOI: 10.1186/s12889-024-20008-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Black men consistently have higher rates of prostate cancer (PCA)- related mortality. Advances in PCA treatment, screening, and hereditary cancer assessment center around germline testing (GT). Of concern is the significant under-engagement of Black males in PCA GT, limiting the benefit of precision therapy and tailored cancer screening despite longstanding awareness of these disparities. To address these critical disparities, the Socioecological Model (SEM) was employed to develop comprehensive recommendations to overcome barriers and implement equitable strategies to engage Black males in PCA GT. METHODS Clinical/research experts, national organization leaders, and community stakeholders spanning multiple regions in US and Africa participated in developing a framework for equity in PCA GT grounded in the SEM. A novel mixed-methods approach was employed to generate key areas to be addressed and informed statements for consensus consideration utilizing the modified Delphi model. Statements achieving strong consensus (> =75% agreement) were included in final equity frameworks addressing clinical/community engagement and research engagement. RESULTS All societal levels of the SEM (interpersonal, institutional, community, and policy/advocacy) must deliver information about PCA GT to Black males that address benefits/limitations, clinical impact, hereditary cancer implications, with acknowledgment of mistrust (mean scores [MS] 4.57-5.00). Interpersonal strategies for information delivery included engagement of family/friends/peers/Black role models to improve education/awareness and overcome mistrust (MS 4.65-5.00). Institutional strategies included diversifying clinical, research, and educational programs and integrating community liaisons into healthcare institutions (MS 4.57-5.00). Community strategies included partnerships with healthcare institutions and visibility of healthcare providers/researchers at community events (MS 4.65-4.91). Policy/advocacy included improving partnerships between advocacy and healthcare/community organizations while protecting patient benefits (MS 4.57-5.00). Media strategies were endorsed for the first time at every level (MS 4.56-5.00). CONCLUSION The SEM-based equity frameworks proposed provide the first multidisciplinary strategies dedicated to increase engagement of Black males in PCA GT, which are critical to reduce disparities in PCA-mortality through informing tailored screening, targeted therapy, and cascade testing in families.
Collapse
Affiliation(s)
- Amy E Leader
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Timothy R Rebbeck
- Department of Epidemiology, Harvard TH Chan School of Public Health and Dana-Farber Cancer Institute, Boston, MA, USA
| | - William K Oh
- Department of Internal Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Alpa V Patel
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Eric P Winer
- Department of Medicine, Yale Cancer Center and Yale School of Medicine, 333 Cedar Street, WWW214A, New Haven, CT, 06520, USA
| | - LeeAnn O Bailey
- National Cancer Institute/Center to Reduce Cancer Health Disparities, Rockville, MD, USA
| | - Leonard G Gomella
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Crystal Y Lumpkins
- Department of Communication, Population Sciences Division, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Isla P Garraway
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Lisa B Aiello
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Monica L Baskin
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Heather H Cheng
- Department of Medicine, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - Kathleen A Cooney
- Department of Medicine, Duke University School of Medicine and Duke Cancer Institute, Durham, NC, USA
| | - Amanda Ganzak
- Cancer Genetics and Prevention Program, Yale New Haven Hospital, New Haven, CT, USA
| | - Daniel J George
- Department of Medicine, Duke University School of Medicine and Duke Cancer Institute, Durham, NC, USA
| | - Susan Halabi
- Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Feighanne Hathaway
- Department of Medicine, High-Risk and Advanced Prostate Cancer Clinic, University of Chicago Medicine, University of Chicago, Chicago, IL, USA
| | - Claire Healy
- Cancer Genetics and Prevention Program, Yale New Haven Hospital, New Haven, CT, USA
| | - Joseph W Kim
- Department of Medicine, Yale Cancer Center and Yale School of Medicine, 333 Cedar Street, WWW214A, New Haven, CT, 06520, USA
| | | | - Stacy Loeb
- Department of Urology and Population Health, New York University and Manhattan Veterans Affairs, New York, NY, USA
| | - Kara N Maxwell
- Department of Medicine-Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher McNair
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Todd M Morgan
- Department of Urology, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Breanne Prindeville
- Neaman Center for Personalized Medicine, NorthShore University Health System, Evanston, IL, USA
| | | | - Whitney L Steward
- National Cancer Institute/Center to Reduce Cancer Health Disparities, Rockville, MD, USA
| | - Sakinah C Suttiratana
- Department of Medicine, Yale Cancer Center and Yale School of Medicine, 333 Cedar Street, WWW214A, New Haven, CT, 06520, USA
| | - Mary-Ellen Taplin
- Department of Medicine, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kosj Yamoah
- Departmetnt of Radiation Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Thierry Fortune
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Joshua Blanding-Godbolt
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Laura Gross
- Department of Medicine, Yale University and Yale Cancer Center, New Haven, CT, USA
| | - Veda N Giri
- Department of Medicine, Yale Cancer Center and Yale School of Medicine, 333 Cedar Street, WWW214A, New Haven, CT, 06520, USA.
| |
Collapse
|
49
|
Klatte DCF, Weston A, Ma Y, Sledge H, Bali A, Bolan C, Engels M, van Hooft JE, van Leerdam ME, Ouni A, Wallace MB, Bi Y. Temporal Trends in Body Composition and Metabolic Markers Prior to Diagnosis of Pancreatic Ductal Adenocarcinoma. Clin Gastroenterol Hepatol 2024; 22:1830-1838.e9. [PMID: 38703880 DOI: 10.1016/j.cgh.2024.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND & AIMS Changes in body composition and metabolic factors may serve as biomarkers for the early detection of pancreatic ductal adenocarcinoma (PDAC). The aim of this study was to capture the longitudinal changes in body composition and metabolic factors before diagnosis of PDAC. METHODS We performed a retrospective cohort study in which all patients (≥18 years) diagnosed with PDAC from 2002 to 2021 were identified. We collected all abdominal computed tomography scans and 10 different blood-based biomarkers up to 36 months before diagnosis. We applied a fully automated abdominal segmentation algorithm previously developed by our group for 3-dimensional quantification of body composition on computed tomography scans. Longitudinal trends of body composition and blood-based biomarkers before PDAC diagnosis were estimated using linear mixed models, compared across different time windows, and visualized using spline regression. RESULTS We included 1690 patients in body composition analysis, of whom 516 (30.5%) had ≥2 prediagnostic computed tomography scans. For analysis of longitudinal trends of blood-based biomarkers, 3332 individuals were included. As an early manifestation of PDAC, we observed a significant decrease in visceral and subcutaneous adipose tissue (β = -1.94 [95% confidence interval (CI), -2.39 to -1.48] and β = -2.59 [95% CI, -3.17 to -2.02]) in area (cm2)/height (m2) per 6 months closer to diagnosis, accompanied by a decrease in serum lipids (eg, low-density lipoprotein [β = -2.83; 95% CI, -3.31 to -2.34], total cholesterol [β = -2.69; 95% CI, -3.18 to -2.20], and triglycerides [β = -1.86; 95% CI, -2.61 to -1.11]), and an increase in blood glucose levels. Loss of muscle tissue and bone volume was predominantly observed in the last 6 months before diagnosis. CONCLUSIONS This study identified significant alterations in a variety of soft tissue and metabolic markers that occur in the development of PDAC. Early recognition of these metabolic changes may provide an opportunity for early detection.
Collapse
Affiliation(s)
- Derk C F Klatte
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Alexander Weston
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Yaohua Ma
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Hanna Sledge
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Aman Bali
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | - Candice Bolan
- Department of Radiology, Mayo Clinic, Jacksonville, Florida
| | - Megan Engels
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeanin E van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands; Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ahmed Ouni
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | - Michael B Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | - Yan Bi
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
50
|
Cheng HH, Shevach JW, Castro E, Couch FJ, Domchek SM, Eeles RA, Giri VN, Hall MJ, King MC, Lin DW, Loeb S, Morgan TM, Offit K, Pritchard CC, Schaeffer EM, Szymaniak BM, Vassy JL, Katona BW, Maxwell KN. BRCA1, BRCA2, and Associated Cancer Risks and Management for Male Patients: A Review. JAMA Oncol 2024; 10:1272-1281. [PMID: 39052257 DOI: 10.1001/jamaoncol.2024.2185] [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] [Indexed: 07/27/2024]
Abstract
Importance Half of all carriers of inherited cancer-predisposing variants in BRCA1 and BRCA2 are male, but the implications for their health are underrecognized compared to female individuals. Germline variants in BRCA1 and BRCA2 (also known as pathogenic or likely pathogenic variants, referred to here as BRCA1/2 PVs) are well known to significantly increase the risk of breast and ovarian cancers in female carriers, and knowledge of BRCA1/2 PVs informs established cancer screening and options for risk reduction. While risks to male carriers of BRCA1/2 PVs are less characterized, there is convincing evidence of increased risk for prostate cancer, pancreatic cancer, and breast cancer in males. There has also been a rapid expansion of US Food and Drug Administration-approved targeted cancer therapies, including poly ADP ribose polymerase (PARP) inhibitors, for breast, pancreatic, and prostate cancers associated with BRCA1/2 PVs. Observations This narrative review summarized the data that inform cancer risks, targeted cancer therapy options, and guidelines for early cancer detection. It also highlighted areas of emerging research and clinical trial opportunities for male BRCA1/2 PV carriers. These developments, along with the continued relevance to family cancer risk and reproductive options, have informed changes to guideline recommendations for genetic testing and strengthened the case for increased genetic testing for males. Conclusions and Relevance Despite increasing clinical actionability for male carriers of BRCA1/2 PVs, far fewer males than female individuals undergo cancer genetic testing. Oncologists, internists, and primary care clinicians should be vigilant about offering appropriate genetic testing to males. Identifying more male carriers of BRCA1/2 PVs will maximize opportunities for cancer early detection, targeted risk management, and cancer treatment for males, along with facilitating opportunities for risk reduction and prevention in their family members, thereby decreasing the burden of hereditary cancer.
Collapse
Affiliation(s)
- Heather H Cheng
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Medicine (Hematology and Oncology), University of Washington, Seattle
| | - Jeffrey W Shevach
- Division of Medical Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Elena Castro
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fergus J Couch
- Division of Experimental Pathology and Laboratory Medicine, Mayo Clinic, Rochester, New York
| | - Susan M Domchek
- Department of Medicine, Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Rosalind A Eeles
- The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Veda N Giri
- Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut
| | - Michael J Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Mary-Claire King
- Department of Medicine (Medical Genetics) and Department of Genome Sciences, University of Washington, Seattle
| | - Daniel W Lin
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Urology, University of Washington, Seattle
| | - Stacy Loeb
- Department of Urology and Population Health, New York University School of Medicine, New York
- Department of Surgery/Urology, Manhattan Veterans Affairs, New York, New York
| | - Todd M Morgan
- Department of Urology, University of Michigan, Ann Arbor
| | - Kenneth Offit
- Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Colin C Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
- Brotman Baty Institute for Precision Medicine, Seattle, Washington
| | - Edward M Schaeffer
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Brittany M Szymaniak
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jason L Vassy
- Harvard Medical School at VA Boston Healthcare System, Boston, Massachusetts
| | - Bryson W Katona
- Department of Medicine, Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Kara N Maxwell
- Department of Medicine, Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Philadelphia
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania
| |
Collapse
|