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
|
He R, Shen Z, Chen Q, Hu H, Ding X, Zheng Z, Feng Q, Li B. Pancreatic cancer mortality in China from 2004 to 2021: an in-depth analysis of age, gender, and regional disparities. BMC Cancer 2025; 25:891. [PMID: 40389880 PMCID: PMC12087137 DOI: 10.1186/s12885-025-13863-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/04/2025] [Indexed: 05/21/2025] Open
Abstract
OBJECTIVE This study aimed to analyze the trends and epidemiological characteristics of pancreatic cancer (PC) mortality in China from 2004 to 2021, focusing on gender, age, and regional disparities. The goal was to provide a comprehensive understanding of PC mortality and identify key risk factors to support future prevention and control strategies. METHODS Using data from the national Disease Surveillance Point (DSP) system, which covers a large and representative sample of the Chinese population, the study examined pancreatic cancer mortality trends across different age groups, sexes, and regions. Statistical analyses, including the independent-sample t-test and age-period-cohort (APC) model, were employed to assess mortality differences and annual percentage changes from 2004 to 2021. RESULTS The study recorded a significant increase in pancreatic cancer mortality, particularly among males and older adults. Mortality was consistently higher in urban areas, but the growth rate in rural areas surpassed that of urban areas. Regional disparities were also observed, with the eastern region showing the highest mortality rates but slower increases compared to the central and western regions. Key risk factors, including aging, diabetes, smoking, and chronic pancreatitis, were identified, with gender-specific differences linked to lifestyle factors such as smoking and alcohol consumption. CONCLUSION Pancreatic cancer mortality in China has shown significant increases over the past 18 years, especially among males, older adults, and rural populations. The findings highlight the urgent need for targeted public health interventions to address gender- and age-specific risks, as well as healthcare access inequalities in less developed regions. Future research should focus on gathering more granular, individual-level data to better understand the complex interplay of risk factors and inform more effective prevention and treatment strategies.
Collapse
Affiliation(s)
- Rui He
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhengnan Shen
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiuping Chen
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Haiyang Hu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xin Ding
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhenglong Zheng
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Quansheng Feng
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Baixue Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Peduzzi G, Archibugi L, Farinella R, de Leon Pisani RP, Vodickova L, Vodicka P, Kraja B, Sainz J, Bars-Cortina D, Daniel N, Silvestri R, Uysal-Onganer P, Landi S, Dulińska-Litewka J, Comandatore A, Campa D, Hughes DJ, Rizzato C. The exposome and pancreatic cancer, lifestyle and environmental risk factors for PDAC. Semin Cancer Biol 2025; 113:100-129. [PMID: 40368260 DOI: 10.1016/j.semcancer.2025.05.004] [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: 01/17/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/16/2025]
Abstract
Pancreatic cancer (PC), particularly pancreatic ductal adenocarcinoma (PDAC), is a significant global health issue with high mortality rates. PDAC, though only 3 % of cancer diagnoses, causes 7 % of cancer deaths due to its severity and asymptomatic early stages. Risk factors include lifestyle choices, environmental exposures, and genetic predispositions. Conditions like new-onset type 2 diabetes and chronic pancreatitis also contribute significantly. Modifiable risk factors include smoking, alcohol consumption, non-alcoholic fatty pancreatic disease (NAFPD), and obesity. Smoking and heavy alcohol consumption increase PC risk, while NAFPD and obesity, particularly central adiposity, contribute through chronic inflammation and insulin resistance. Refined sugar and sugar-sweetened beverages (SSBs) are also linked to increased PC risk, especially among younger individuals. Hormonal treatments and medications like statins, aspirin, and metformin have mixed results on PC risk, with some showing protective effects. The gut microbiome influences PC through the gut-pancreas axis, with disruptions leading to inflammation and carcinogenesis. Exposure to toxic substances, including heavy metals and chemicals, is associated with increased PC risk. Glycome changes, such as abnormal glycosylation patterns, are significant in PDAC development and offer potential for early diagnosis. Interactions between environmental and genetic factors are crucial in PDAC susceptibility. Genome-wide association studies (GWAS) have identified several single nucleotide polymorphisms (SNPs) linked to PDAC, but gene-environment interactions remain largely unexplored. Future research should focus on polygenic risk scores (PRS) and large-scale studies to better understand these interactions and their impact on PDAC risk.
Collapse
Affiliation(s)
| | - Livia Archibugi
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Ruggero Ponz de Leon Pisani
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Ludmila Vodickova
- Biomedical Center Martin, Bioinformatic Center, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Slovakia; Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Vodicka
- Biomedical Center Martin, Bioinformatic Center, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Slovakia; Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czech Republic
| | - Bledar Kraja
- University Clinic of Gastrohepatology, University Hospital Center Mother Teresa, Tirana, Albania
| | - Juan Sainz
- Department of Biochemistry and Molecular Biology, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid 28029, Spain; GENYO. Centre for Genomics and Oncological Research. Genomic Oncology department, Granada, Spain; Instituto de Investigación Biosanitaria Ibs.Granada, Granada, Spain
| | - David Bars-Cortina
- Institut Català d'Oncologia (ICO) IDIBELL, Unit of Biomarkers and Susceptibility (UBS), Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain; Institut Català d'Oncologia (ICO) IDIBELL, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | | | - Pinar Uysal-Onganer
- Cancer Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London, UK
| | - Stefano Landi
- Department of Biology, University of Pisa, Pisa, Italy
| | | | - Annalisa Comandatore
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | | |
Collapse
|
4
|
Quammie S, Crooks CJ, Aliyu A, Aithal GP, Aravinthan AD. Chronic pancreatitis and extra pancreatic cancers- A systematic review and meta analysis. Pancreatology 2025:S1424-3903(25)00088-2. [PMID: 40382256 DOI: 10.1016/j.pan.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 03/11/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025]
Abstract
INTRODUCTION Chronic pancreatitis (CP) is a known risk factor for pancreatic cancer; however its association to extra pancreatic (EP) cancers remains inadequately explored. The aim of this systematic review is to investigate the evidence for CP as a risk factor for developing EP cancers. METHOD Electronic search was conducted on Ovid Medline, EMBASE and Scopus from inception to January 27, 2024 to identify patients with CP who developed EP cancers. Prevalence and incidence of each cancer were calculated where possible from the reported numbers. A random effects meta-analysis was used to pool prevalence, incidence and hazard ratios (HR) of each EP cancer. Heterogeneity was assessed using I2. PROSPERO registration: CRD42024543050. RESULTS Sixteen (16) studies consisting of 117,163 CP patients met the eligibility criteria. 4015 (3.4%) patients developed 4019 EP cancers. The overall annual prevalence and incidence of EP cancers were 7962 (95% CI 5044-10880) per 100,000 CP patients and 1039 (95% CI 649-1663) per 100,000 person years. Lung cancer had the highest annual prevalence - (1540 (95% CI 667-2413) per 100,000 CP patients) and incidence (260 (95% CI 120-390) per 100,000 person years). The pooled HR were 1.31 (95% CI 1.03-1.66) and 1.58 (95% CI 1.27-1.98) for adjusted lung cancer and crude liver cancer in patients with CP compared to patients without CP, respectively. CONCLUSION Patients with CP have an increased risk of developing EP cancers compared to patients without CP, in particular lung and liver cancer which had the highest relative risk. Shared risk factor modifications, such as smoking cessation and alcohol reduction, could lower the risk of common EP cancers. Further, implementing non-invasive screening measures may aid in early diagnosis in this high-risk group.
Collapse
Affiliation(s)
- Shauntelle Quammie
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, UK; Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Colin John Crooks
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, UK; Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Abdulsalam Aliyu
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Guruprasad P Aithal
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, UK; Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Aloysious D Aravinthan
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, UK; Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK.
| |
Collapse
|
5
|
Seidel T, Ohri N, Glaß M, Sunami Y, Müller LP, Kleeff J. Stromal Cells in Early Inflammation-Related Pancreatic Carcinogenesis-Biology and Its Potential Role in Therapeutic Targeting. Cancers (Basel) 2025; 17:1541. [PMID: 40361466 DOI: 10.3390/cancers17091541] [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: 04/03/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
The stroma of healthy pancreases contains various non-hematopoietic, non-endothelial mesenchymal cells. It is altered by chronic inflammation which in turn is a major contributor to the development of pancreatic adenocarcinoma (PDAC). In PDAC, the stroma plays a decisive and well-investigated role for tumor progression and therapy response. This review addresses the central role of stromal cells in the early inflammation-driven development of PDAC. It focuses on major subpopulations of pancreatic mesenchymal cells, i.e., fibroblasts, pancreatic stellate cells, and multipotent stroma cells, particularly their activation and functional alterations upon chronic inflammation including the development of different types of carcinoma-associated fibroblasts. In the second part, the current knowledge on the impact of activated stroma cells on acinar-to-ductal metaplasia and the transition to pancreatic intraepithelial neoplasia is summarized. Finally, putative strategies to target stroma cells and their signaling in early pancreatic carcinogenesis are reflected. In summary, the current data show that the activation of pancreatic stroma cells and the resulting fibrotic changes has pro- and anti-carcinogenetic effects but, overall, creates a carcinogenesis-promoting microenvironment. However, this is a dynamic process and the therapeutic targeting of specific pathways and cells requires in-depth knowledge of the molecular interplay of various cell types.
Collapse
Affiliation(s)
- Tina Seidel
- Department of Internal Medicine, University Hospital Halle, 06120 Halle (Saale), Germany
| | - Nupur Ohri
- Department of Visceral, Vascular and Endocrine Surgery, University Hospital Halle, 06120 Halle (Saale), Germany
| | - Markus Glaß
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany
| | - Yoshiaki Sunami
- Department of Visceral, Vascular and Endocrine Surgery, University Hospital Halle, 06120 Halle (Saale), Germany
| | - Lutz P Müller
- Department of Internal Medicine, University Hospital Halle, 06120 Halle (Saale), Germany
| | - Jörg Kleeff
- Department of Visceral, Vascular and Endocrine Surgery, University Hospital Halle, 06120 Halle (Saale), Germany
| |
Collapse
|
6
|
Liang Y, Sui X, Li S, Peng H, Jiang W, Jia M, Jiang S, Wang W, Teng H. Development and validation of a predictive model based upon extracellular vesicle-derived transposable elements for non-invasive detection of pancreatic adenocarcinoma. Biomark Res 2025; 13:54. [PMID: 40188348 PMCID: PMC11972517 DOI: 10.1186/s40364-025-00770-6] [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: 10/22/2024] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
Pancreatic adenocarcinoma (PAAD) is a highly lethal malignancy that leads to patients missing optimal treatment opportunities due to its atypical clinical symptoms and the lack of effective diagnostic biomarkers. To develop a biomarker panel based on extracellular vesicle-derived transposable elements (EV-TEs) for non-invasive detection of PAAD, we analyzed 6.75 Tbp sequencing data of 852 EV-derived transcriptomes from two cohorts, and identified 31 EV-TEs features as the biomarker panel using recursive feature elimination. Predictive model constructed using the Support Vector Machine (SVM) algorithm demonstrated excellent performance for PAAD detection in the training set (AUC: 0.90, 95% CI: 0.86-0.93), the test set (AUC: 0.86, 95% CI: 0.79-0.92) and the independent external validation cohort of blood EV-derived samples (AUC: 0.88, 95% CI: 0.84-0.92). This study presents the first EV-TEs based predictive model for PAAD detection, showcasing the immense potential of these 'junk DNA' as innovative diagnostic biomarker for cancers.
Collapse
Affiliation(s)
- Yueting Liang
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xin Sui
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Shuai Li
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Haoxin Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Wenyi Jiang
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Minqi Jia
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Shaoran Jiang
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Weihu Wang
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Huajing Teng
- Department of Radiation Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
7
|
Wu H, Gao SZ, Yin LD, Wang YH, Deng SJ, Lu ZP, Jin G, Zou WB. Germline mutations and pancreatic cancer risk in chronic pancreatitis. Gut 2025:gutjnl-2024-334277. [PMID: 40187890 DOI: 10.1136/gutjnl-2024-334277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/19/2025] [Indexed: 04/07/2025]
Affiliation(s)
- Hao Wu
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Sui-Zhi Gao
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Ling-Di Yin
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ya-Hui Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Shun-Jiang Deng
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zi-Peng Lu
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Wen-Bin Zou
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
8
|
Zhu W, Chen L, Aphinyanaphongs Y, Kastrinos F, Simeone DM, Pochapin M, Stender C, Razavian N, Gonda TA. Identification of patients at risk for pancreatic cancer in a 3-year timeframe based on machine learning algorithms. Sci Rep 2025; 15:11697. [PMID: 40188106 PMCID: PMC11972345 DOI: 10.1038/s41598-025-89607-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 02/06/2025] [Indexed: 04/07/2025] Open
Abstract
Early detection of pancreatic cancer (PC) remains challenging largely due to the low population incidence and few known risk factors. However, screening in at-risk populations and detection of early cancer has the potential to significantly alter survival. In this study, we aim to develop a predictive model to identify patients at risk for developing new-onset PC at two and a half to three year time frame. We used the Electronic Health Records (EHR) of a large medical system from 2000 to 2021 (N = 537,410). The EHR data analyzed in this work consists of patients' demographic information, diagnosis records, and lab values, which are used to identify patients who were diagnosed with pancreatic cancer and the risk factors used in the machine learning algorithm for prediction. We identified 73 risk factors of pancreatic cancer with the Phenome-wide Association Study (PheWAS) on a matched case-control cohort. Based on them, we built a large-scale machine learning algorithm based on EHR. A temporally stratified validation based on patients not included in any stage of the training of the model was performed. This model showed an AUROC at 0.742 [0.727, 0.757] which was similar in both the general population and in a subset of the population who has had prior cross-sectional imaging. The rate of diagnosis of pancreatic cancer in those in the top 1 percentile of the risk score was 6 folds higher than the general population. Our model leverages data extracted from a 6-month window of time in the electronic health record to identify patients at nearly sixfold higher than baseline risk of developing pancreatic cancer 2.5-3 years from evaluation. This approach offers an opportunity to define an enriched population entirely based on static data, where current screening may be recommended.
Collapse
Affiliation(s)
- Weicheng Zhu
- Center for Data Science, New York University, New York, NY, USA
| | - Long Chen
- Center for Data Science, New York University, New York, NY, USA
| | - Yindalon Aphinyanaphongs
- Department of Population Health, New York University Grossman School of Medicine, 227 East 30th Street, 6th Floor, New York, NY, 10016, USA
| | - Fay Kastrinos
- Department of Medicine, Division of Digestive and Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Diane M Simeone
- Moores Cancer Center, UC San Diego Health, San Diego, CA, USA
| | - Mark Pochapin
- Division of Gastroenterology and Hepatology, Department of Medicine, New York University, 240 East 38th Street, 23rd Floor, New York, NY, 10016, USA
| | - Cody Stender
- Department of Surgery, New York University, New York, NY, USA
| | - Narges Razavian
- Department of Population Health, New York University Grossman School of Medicine, 227 East 30th Street, 6th Floor, New York, NY, 10016, USA.
| | - Tamas A Gonda
- Division of Gastroenterology and Hepatology, Department of Medicine, New York University, 240 East 38th Street, 23rd Floor, New York, NY, 10016, USA.
| |
Collapse
|
9
|
Paquette M, Guay SP, Baass A. Genetic determinants of pancreatitis risk in hypertriglyceridemia. Curr Opin Lipidol 2025; 36:55-60. [PMID: 39513935 DOI: 10.1097/mol.0000000000000962] [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] [Indexed: 11/16/2024]
Abstract
PURPOSE OF REVIEW In recent years, studies have shed light on the concept of risk heterogeneity among patients with severe hypertriglyceridemia (HTG). Several clinical risk factors for acute pancreatitis have been identified in this population, but the importance of different genetic factors above and beyond triglyceride concentration remains unclear. This review endeavours to summarize recent developments in this field. RECENT FINDINGS Recent studies suggest that the molecular basis of severe HTG (polygenic susceptibility vs. rare pathogenic variants) can modulate the risk of acute pancreatitis independently of triglyceride level. Furthermore, a pancreatitis polygenic risk score has been developed and validated using data from the largest GWAS meta-analysis of acute pancreatitis published to date. In patients with severe HTG, a high polygenic susceptibility for pancreatitis was associated with a three-fold increased risk of acute pancreatitis compared with those with a lower polygenic risk score. SUMMARY In the past months, there have been substantial advances in understanding the prediction of acute pancreatitis in patients with severe HTG. However, further efforts at developing risk-stratification strategies and predictive models may help identifying the patients who would benefit most from early and effective interventions to reduce the risk of pancreatitis, including treatment with APOC3 inhibitors.
Collapse
Affiliation(s)
- Martine Paquette
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montreal
| | - Simon-Pierre Guay
- Division of Medical Genetics, Department of Pediatric, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke
| | - Alexis Baass
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montreal
- Department of Medicine, Divisions of Experimental Medicine and Medical Biochemistry, McGill University, Montreal, Québec, Canada
| |
Collapse
|
10
|
Sharma R, Ghosh R, Kumar S, Komal K, Kumar M. Decoding pancreatic cancer: key risk factors across genetics and lifestyle. Expert Rev Mol Diagn 2025; 25:95-99. [PMID: 39918844 DOI: 10.1080/14737159.2025.2464563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/05/2025] [Indexed: 02/09/2025]
Affiliation(s)
- Rohit Sharma
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Rashmi Ghosh
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Kumari Komal
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College Pharmacy, Moga, India
| |
Collapse
|
11
|
Han H, Chen BT, Liu Y, Qi L, Xing L, Wang H, Zhao M, Zhang C, Yu P, Wei N, Wang J, Zhou F, Wang GJ, Cheng XW, Huang ZJ, Li L, Jiang HL. Engineered Stem Cell Booster Breaks Pathological Barriers to Treat Chronic Pancreatitis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2416261. [PMID: 40012418 DOI: 10.1002/adma.202416261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/26/2025] [Indexed: 02/28/2025]
Abstract
Chronic pancreatitis (CP) is a long-standing progressive fibrosis and has long been considered incurable, which remains a heavy health burden worldwide. Mesenchymal stem cells (MSCs) with anti-fibrosis properties are currently used in the treatment of fibroinflammatory diseases. However, its therapeutic effect is limited mainly due to two main types of pathological barriers in CP: 1) Fibrotic collagen hinders cell delivery, and 2) Malignant microenvironment attacks cell inactivation. Here, a MSCs-based exogenous nitric oxide (NO) delivery system (MSCs-Lip@RNO) is constructed. In the MSCs-Lip@RNO, NO not only can be a cell booster to regulate collagen fibers, relieve the vascular compression and enhance the accumulation of MSCs in the whole pancreas, but also can form a protective gas layer on the cell surface, which enhances the therapeutic effect of MSCs. In the CP rat model, the pancreatic injury and fibrosis are reduced with 7 days after a single dose administration of this long-acting MSCs. Collectively, this study offers a promising strategy for enhancing the delivery and therapeutic efficacy of MSCs to break pathological barriers in CP treatment.
Collapse
Affiliation(s)
- Han Han
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Bi-Te Chen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Liang Qi
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, P. R. China
- College of Pharmacy, Yanbian University, Yanji, 133002, P. R. China
| | - Hui Wang
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Min Zhao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Chen Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Ping Yu
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Ning Wei
- Jiangsu Renocell Biotech Co., Ltd, Nanjing, 211100, P. R. China
| | - Jing Wang
- Jiangsu Renocell Biotech Co., Ltd, Nanjing, 211100, P. R. China
| | - Fang Zhou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Guang-Ji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Xian-Wu Cheng
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, P. R. China
| | - Zhang-Jian Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing, 210009, P. R. China
- School of Pharmacy, Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology, Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Xinjiang Medical University, Urumqi, 830054, P. R. China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, P. R. China
- College of Pharmacy, Yanbian University, Yanji, 133002, P. R. China
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
12
|
Damiano OM, Stevens AJ, Kenwright DN, Seddon AR. Chronic Inflammation to Cancer: The Impact of Oxidative Stress on DNA Methylation. FRONT BIOSCI-LANDMRK 2025; 30:26142. [PMID: 40152377 DOI: 10.31083/fbl26142] [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: 08/15/2024] [Revised: 11/10/2024] [Accepted: 11/21/2024] [Indexed: 03/29/2025]
Abstract
The genomic landscape of cancer cells is complex and heterogeneous, with aberrant DNA methylation being a common observation. Growing evidence indicates that oxidants produced from immune cells may interact with epigenetic processes, and this may represent a mechanism for the initiation of altered epigenetic patterns observed in both precancerous and cancerous cells. Around 20% of cancers are linked to chronic inflammatory conditions, yet the precise mechanisms connecting inflammation with cancer progression remain unclear. During chronic inflammation, immune cells release oxidants in response to stimuli, which, in high concentrations, can cause cytotoxic effects. Oxidants are known to damage DNA and proteins and disrupt normal signalling pathways, potentially initiating a sequence of events that drives carcinogenesis. While research on the impact of immune cell-derived oxidants on DNA methylation remains limited, this mechanism may represent a crucial link between chronic inflammation and cancer development. This review examines current evidence on inflammation-associated DNA methylation changes in cancers related to chronic inflammation.
Collapse
Affiliation(s)
- Olivia M Damiano
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Aaron J Stevens
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Diane N Kenwright
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Annika R Seddon
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, 8011 Christchurch, New Zealand
| |
Collapse
|
13
|
Cheng H, Guo H, Wen C, Sun G, Tang F, Li Y. The dual role of gut microbiota in pancreatic cancer: new insights into onset and treatment. Ther Adv Med Oncol 2025; 17:17588359251324882. [PMID: 40093983 PMCID: PMC11909682 DOI: 10.1177/17588359251324882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Pancreatic cancer ranks among the most lethal digestive malignancies, exhibiting a steadily increasing incidence and mortality worldwide. Despite significant advances in cancer research, the 5-year survival rate remains below 10%, predominantly due to delayed diagnosis and limited therapeutic options. Concurrently, the gut microbiota-an integral component of host physiology-has emerged as a crucial player in the pathogenesis of pancreatic cancer. Mounting evidence indicates that alterations in gut microbial composition and function may influence tumor initiation, progression, and response to therapy. This review provides an in-depth examination of the intricate interplay between the gut microbiome and pancreatic cancer, highlighting potential diagnostic biomarkers and exploring microbiome-targeted therapeutic strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Huijuan Cheng
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu, P.R. China
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, P.R. China
| | - Hongkai Guo
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
| | - Chengming Wen
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
| | - Guodong Sun
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
- Department of Medical Affairs, Lanzhou University First Hospital, Lanzhou, Gansu, P.R. China
| | - Futian Tang
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, P.R. China
| | - Yumin Li
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
14
|
Bampton TJ, Chen JW, Brown A, Barnett MI, Coates PT, Palmer LJ. Epidemiology and burden of adult chronic pancreatitis in South Australia: a 20-year data linkage study. BMJ Open 2025; 15:e089297. [PMID: 40050052 PMCID: PMC11887304 DOI: 10.1136/bmjopen-2024-089297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 01/24/2025] [Indexed: 03/09/2025] Open
Abstract
OBJECTIVES To investigate the epidemiology and burden of adult-onset chronic pancreatitis (CP) in South Australia. DESIGN Retrospective case-control study; data linkage. SETTING All public adult hospitals in SA. PARTICIPANTS Administrative data linkage from South Australia-Northern Territory DataLink was used to ascertain an index cohort of all adults with an initial diagnosis of CP aged >19 years between June 2000 and June 2019. Age- and sex-matched controls were drawn from the general population of SA, adults with type 1 diabetes mellitus and adults with type 2 diabetes mellitus (defined by International Classification of Diseases 10th Revision coding). MAIN OUTCOME MEASURES Hospital visits, days in hospital, emergency department visits, intensive care unit admissions, incidence, prevalence. RESULTS A total of 2503 incident index cases with CP were identified. The crude prevalence and incidence were estimated as 195.1 per 100 000 and 10.4 per 100 000 per annum, respectively. Cases of CP averaged more hospital visits for any reason (median 11, IQR 5 to 21.75) than the general population (median 1, IQR 0 to 4) and had a higher healthcare burden than controls with type 1 diabetes or type 2 diabetes (all p<0.001). Indigenous individuals were over-represented in the cohort (n=358; 14.8% vs 1.5% of the general population) and had higher healthcare utilisation than other patients with CP (p<0.001). CONCLUSIONS CP is a significant burden on the SA healthcare system and was more prevalent and more burdensome in Indigenous adults. CP consumes a disproportionate level of public health services. Our findings support further research and preventive efforts, particularly in the Indigenous population.
Collapse
Affiliation(s)
- Tristan J Bampton
- School of Public Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - John W Chen
- Department of Surgery, Flinders Medical Centre, Bedford Park, Australia
| | - Alex Brown
- Australian National University, Canberra, Australia
| | - Meghan I Barnett
- School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - P Toby Coates
- Central Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, South Australia, Australia
- School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Lyle John Palmer
- School of Public Health, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
15
|
Wei W, Ma Y, Zeng J, Song Y, Han Y, Qian W, Yang X, Wu Z, Ma Z, Wang Z, Duan W. A Nomogram for Predicting the Transition From Recurrent Acute Pancreatitis to Chronic Pancreatitis. Pancreas 2025; 54:e201-e209. [PMID: 39999313 PMCID: PMC11882177 DOI: 10.1097/mpa.0000000000002420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/11/2024] [Indexed: 02/27/2025]
Abstract
OBJECTIVES Acute pancreatitis, recurrent acute pancreatitis, and chronic pancreatitis are recognized as a continuum of pancreatic diseases. Recurrence increases the risk of progression to chronic pancreatitis. The aim of this study was to search for clinical features that may promote the progression of chronic pancreatitis in patients with recurrent acute pancreatitis. MATERIALS AND METHODS We retrospectively reviewed patients with recurrent acute pancreatitis from Medical Information Mart for Intensive Care-IV database. They were divided into a training cohort and a validation cohort. A nomogram was constructed based on clinical features during the second hospitalization. The discrimination and calibration of the nomogram were evaluated using the concordance index, area under the time-dependent receiver operating characteristic curve, and calibration plots. RESULTS A total of 432 recurrent acute pancreatitis patients were evaluated, of which 93 (21.53%) were diagnosed with chronic pancreatitis later. Age, biliary pancreatitis, admission interval, alcohol dependence, lipase, and platelet were selected. The concordance index was 0.717 (95% confidence interval: 0.691-0.743) for the training cohort and 0.718 (95% confidence interval: 0.662-0.774) for the validation cohort. The area under the time-dependent receiver operating characteristic curve was >0.7 over 1000 days. CONCLUSIONS A nomogram was developed and validated to evaluate the transition from recurrent acute pancreatitis to chronic pancreatitis.
Collapse
|
16
|
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
|
17
|
Kim YE, Yu MH, Nam CM, Kang ES. Effects of Pancreatitis and Type 2 Diabetes Mellitus on the Development of Pancreatic Cancer: A Nationwide Nested Case-Control Study. Diabetes Metab J 2025; 49:252-263. [PMID: 40073907 PMCID: PMC11960203 DOI: 10.4093/dmj.2024.0277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/23/2024] [Indexed: 03/14/2025] Open
Abstract
BACKGRUOUND Despite diabetes mellitus (DM) and pancreatitis being known risk factors for pancreatic cancer, patients with these conditions are not included in pancreatic cancer screening due to the low incidence of pancreatic cancer in these populations. This study aimed to determine the high-risk subgroup of patients with diabetes and pancreatitis that would benefit from pancreatic cancer screening. METHODS A nested case-control study was conducted using data from the National Health Information Database of the Korean National Health Insurance Service. Patients were categorized into the following groups: type 2 diabetes mellitus only (T2DM-only), pancreatitis-only (PAN-only), T2DM followed by pancreatitis (T2DM-PAN), post-pancreatitis diabetes mellitus (PPDM), and no diabetes and no pancreatitis (NDNP). Conditional logistic regression was used to determine significant associations of each group with pancreatic cancer development risk. RESULTS The risk of pancreatic cancer was significantly higher in the T2DM-PAN (adjusted odds ratio [AOR], 4.96; 95% confidence interval [CI], 4.48 to 5.49) and PPDM (AOR, 4.71; 95% CI, 4.12 to 5.37) groups than in the NDNP group. Compared to patients in the NDNP group, those with PPDM using insulin had a 17-fold increased risk (AOR, 16.72; 95% CI, 9.50 to 29.43), and individuals with PPDM who had diabetes for less than 3 years had a more than 8-fold increased risk of pancreatic cancer (AOR, 8.83; 95% CI, 5.99 to 13.01). CONCLUSION In patients with post-pancreatitis diabetes, insulin use or shorter duration of diabetes was associated with a higher risk of pancreatic cancer, suggesting that patients in these subgroups may require close monitoring for pancreatic cancer development.
Collapse
Affiliation(s)
- Young-eun Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Min Heui Yu
- SENTINEL Team, Division of Endocrinology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Chung Mo Nam
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institute of Health Services Research, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Seok Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Vujasinovic M, Ebrahimi F, Roelstraete B, Bergman D, Sun J, Sadr‐Azodi O, Löhr J, Ludvigsson JF. Metabolic Dysfunction-Associated Steatotic Liver Disease and Pancreatic Disease-A Population-Based Nationwide Cohort and Sibling-Controlled Study. United European Gastroenterol J 2025; 13:247-256. [PMID: 39868838 PMCID: PMC11975618 DOI: 10.1002/ueg2.12761] [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: 09/24/2024] [Revised: 12/04/2024] [Accepted: 12/19/2024] [Indexed: 01/28/2025] Open
Abstract
INTRODUCTION Metabolic dysfunction-associated steatotic liver disease (MASLD) has been linked to pancreatic diseases, but evidence from population-based studies with liver histology is lacking. AIMS AND METHODS In this population-based cohort including all Swedish adults (n = 8563) with biopsy-proven MASLD, we aimed to investigate incidences of pancreatic diseases compared with matched reference individuals from the general population (n = 38,858) and full siblings (n = 6696). Using Cox proportional hazard models, we calculated multivariable adjusted hazard ratios (aHRs) and confidence intervals (CIs). RESULTS We documented 359 incidents of pancreatic diseases in MASLD patients and 880 events in matched reference individuals, resulting in an incidence rate difference of 1.54 (95% CI, 1.25-1.84). The relative risk of pancreatic disease was highest in the first two years after MASLD diagnosis (aHR, 2.19 [95% CI, 1.92-2.50), but remained statistically significant increased even up to ten years [aHR, 1.60 (95% CI, 1.38-1.85)]. The most common pancreatic disease in individuals with MASLD was acute non-biliary pancreatitis (1.44 vs. 0.44 events/1000 PY), followed by chronic pancreatitis (0.54 vs. 0.12/1000 PY) and pancreatic cancer (0.88 vs. 0.47/1000 PY). We documented 130 versus 344 pancreas-related deaths among individuals with MASLD and their matched comparators, yielding an absolute risk difference of 0.51/1000 PY and an aHR of 2.41 (95%CI = 1.95-2.97). The findings were consistent in sibling-controlled analyses with an aHR of 2.21 (95%CI = 1.69-2.90). CONCLUSIONS MASLD was associated with significantly higher rates of acute and chronic pancreatitis of predominantly non-biliary origin, as well as an increased risk of pancreatic cancer and pancreas-related mortality.
Collapse
Affiliation(s)
- Miroslav Vujasinovic
- Department of Upper Abdominal DiseasesKarolinska University HospitalStockholmSweden
- Department of Medicine HuddingeKarolinska InstitutetStockholmSweden
| | - Fahim Ebrahimi
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Department of Gastroenterology and HepatologyUniversity Digestive Health Care Center Basel—ClarunisBaselSwitzerland
| | - Bjorn Roelstraete
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - David Bergman
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Jiangwei Sun
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Omid Sadr‐Azodi
- Department of Clinical ScienceIntervention and TechnologyKarolinska InstitutetStockholmSweden
- Unit of Upper Gastrointestinal SurgerySaint Göran's HospitalStockholmSweden
- Centre for Clinical Research SörmlandUppsala UniversityEskilstunaSweden
| | - J.‐Matthias Löhr
- Department of Upper Abdominal DiseasesKarolinska University HospitalStockholmSweden
- Department of Clinical ScienceIntervention and TechnologyKarolinska InstitutetStockholmSweden
| | - Jonas F. Ludvigsson
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Celiac Disease CenterDepartment of MedicineColumbia University Medical CenterNew YorkNew YorkUSA
- Department of PediatricsÖrebro University HospitalÖrebro UniversityÖrebroSweden
| |
Collapse
|
19
|
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
|
20
|
Balaban M, Balaban DV, Mănucu G, Bucurică SN, Costache RS, Ioniță-Radu F, Jinga M, Gheorghe C. Groove Pancreatitis in Focus: Tumor-Mimicking Phenotype, Diagnosis, and Management Insights. J Clin Med 2025; 14:1627. [PMID: 40095606 PMCID: PMC11901001 DOI: 10.3390/jcm14051627] [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: 01/27/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Background/Objectives: Groove pancreatitis (GP) is an uncommon pancreatic condition implying a challenging differential diagnosis. This study aims to comprehensively evaluate the main risk factors, clinical presentation, imaging and endoscopic characteristics of patients with GP, providing insights into an effective diagnostic approach and therapeutic strategies. Methods: A retrospective analysis was conducted on patients diagnosed with GP, with demographic and clinical data collected. The diagnostic route was followed by an upper endoscopy and was finally confirmed by cross-sectional imaging. In patients with high malignancy suspicion or with an uncertain diagnosis, a pancreatic endoscopic ultrasound (EUS) was further performed. According to imaging features, we divided patients into two categories: with and without tumor-like appearance. Results: Altogether, 23 patients were included, 11 in the tumor-like category, and 12 in the non-tumor-like group; 95.6% were men, 78.2% alcohol consumers, and 73.9% smokers. In both groups, the main symptom was abdominal pain, followed by nausea and vomiting. The most frequent finding at upper endoscopy was edematous duodenal mucosa (16 patients, 80%), followed by mucosal hyperemia (8 patients, 40%). The main finding at cross-sectional imaging was duodenal wall thickening (14 patients, 60.9%), followed by pancreatic head enlargement and duodenal wall cysts (both seen in 12 patients, 52.2%). The EUS predominantly showed duodenal wall thickening (13 patients, 68.4%), and intramural and paraduodenal cysts (10 patients, 52.6%). Conclusions: GP predominantly affects men with a history of chronic alcohol and tobacco use. Its primary diagnostic challenge lies in distinguishing it from pancreatic carcinoma, with an accurate diagnostic workup being crucial in clinical practice.
Collapse
Affiliation(s)
- Marina Balaban
- Doctoral School, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Daniel Vasile Balaban
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - George Mănucu
- Doctoral School, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Radiology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Săndica Nicoleta Bucurică
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Raluca Simona Costache
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Florentina Ioniță-Radu
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Mariana Jinga
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Cristian Gheorghe
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania
| |
Collapse
|
21
|
Chen X, Sun F, Wang X, Feng X, Aref AR, Tian Y, Ashrafizadeh M, Wu D. Inflammation, microbiota, and pancreatic cancer. Cancer Cell Int 2025; 25:62. [PMID: 39987122 PMCID: PMC11847367 DOI: 10.1186/s12935-025-03673-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/04/2025] [Indexed: 02/24/2025] Open
Abstract
Pancreatic cancer (PC) is a malignancy of gastrointestinal tract threatening the life of people around the world. In spite of the advances in the treatment of PC, the overall survival of this disease in advanced stage is less than 12%. Moreover, PC cells have aggressive behaviour in proliferation and metastasis as well as capable of developing therapy resistance. Therefore, highlighting the underlying molecular mechanisms in PC pathogenesis can provide new insights for its treatment. In the present review, inflammation and related pathways as well as role of gut microbiome in the regulation of PC pathogenesis are highlighted. The various kinds of interleukins and chemokines are able to regulate angiogenesis, metastasis, proliferation, inflammation and therapy resistance in PC cells. Furthermore, a number of molecular pathways including NF-κB, TLRs and TGF-β demonstrate dysregulation in PC aggravating inflammation and tumorigenesis. Therapeutic regulation of these pathways can reverse inflammation and progression of PC. Both chronic and acute pancreatitis have been shown to be risk factors in the development of PC, further highlighting the role of inflammation. Finally, the composition of gut microbiota can be a risk factor for PC development through affecting pathways such as NF-κB to mediate inflammation.
Collapse
Affiliation(s)
- XiaoLiang Chen
- Department of General Surgery and Integrated Traditional Chinese and Western Medicine Oncology, Tiantai People'S Hospital of Zhejiang Province(Tiantai Branch of Zhejiang Provincial People'S Hospital), Hangzhou Medical College, Taizhou, Zhejiang, China
| | - Feixia Sun
- Nursing Department, Shandong First Medical University Affiliated Occupational Disease Hospital (Shandong Provincial Occupational Disease Hospital), Jinan, China
| | - Xuqin Wang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401120, China
| | - Xiaoqiang Feng
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, 525200, Guangdong, China
| | - Amir Reza Aref
- VitroVision Department, DeepkinetiX, Inc, Boston, MA, USA
| | - Yu Tian
- Research Center, the Huizhou Central People'S Hospital, Guangdong Medical University, Huizhou, Guangdong, China.
- School of Public Health, Benedictine University, No. 5700 College Road, Lisle, IL, 60532, USA.
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China.
| | - Dengfeng Wu
- Department of Emergency, The People'S Hospital of Gaozhou, No. 89 Xiguan Road, Gaozhou, 525200, Guangdong, China.
| |
Collapse
|
22
|
Wang X, Yu P, Jia W, Wan B, Ling Z, Tang Y. Integrating traditional machine learning with qPCR validation to identify solid drug targets in pancreatic cancer: a 5-gene signature study. Front Pharmacol 2025; 15:1539120. [PMID: 39850570 PMCID: PMC11754184 DOI: 10.3389/fphar.2024.1539120] [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: 12/03/2024] [Accepted: 12/20/2024] [Indexed: 01/25/2025] Open
Abstract
Background Pancreatic cancer remains one of the deadliest malignancies, largely due to its late diagnosis and lack of effective therapeutic targets. Materials and methods Using traditional machine learning methods, including random-effects meta-analysis and forward-search optimization, we developed a robust signature validated across 14 publicly available datasets, achieving a summary AUC of 0.99 in training datasets and 0.89 in external validation datasets. To further validate its clinical relevance, we analyzed 55 peripheral blood samples from pancreatic cancer patients and healthy controls using qPCR. Results This study identifies and validates a novel five-gene transcriptomic signature (LAMC2, TSPAN1, MYO1E, MYOF, and SULF1) as both diagnostic biomarkers and potential drug targets for pancreatic cancer. The differential expression of these genes was confirmed, demonstrating their utility in distinguishing cancer from normal conditions with an AUC of 0.83. These findings establish the five-gene signature as a promising tool for both early, non-invasive diagnostics and the identification of actionable drug targets. Conclusion A five-gene signature is established robustly and has utility in diagnostics and therapeutic targeting. These findings lay a foundation for developing diagnostic tests and targeted therapies, potentially offering a pathway toward improved outcomes in pancreatic cancer management.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Pengcheng Yu
- Department of General Surgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Wei Jia
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bingbing Wan
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhougui Ling
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Yangyang Tang
- Department of General Surgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| |
Collapse
|
23
|
Assarian BA, Byrne CD, McDonnell D, Hamady Z. Physical Activity and Incident Pancreatic Cancer: Results From the UK Biobank Prospective Cohort. Cureus 2025; 17:e78040. [PMID: 39872920 PMCID: PMC11770282 DOI: 10.7759/cureus.78040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2025] [Indexed: 01/30/2025] Open
Abstract
Background The relationship between physical activity and incident pancreatic cancer is poorly defined, and the evidence to date is inconsistent, largely due to small sample sizes and insufficient incident outcomes. Using the UK Biobank cohort dataset, the association between physical activity levels at recruitment and incident pancreatic ductal adenocarcinoma (PDAC) at follow-up was analysed. Method Physical activity, the key exposure, was quantified using Metabolic Equivalent Task (MET) values and categorised into walking, moderate, and vigorous activity. These categories were each analysed in quartiles. Summed activity was analysed both in quartiles and using International Physical Activity Questionnaire (IPAQ) activity levels (low, moderate, high). Univariate hazard ratios (HRs) and multivariable-adjusted HRs (aHRs) with 95% confidence intervals (CIs) were calculated using Cox regression analyses. Results A total of 542 incident PDAC cases and 2,139 controls (1:4 matching for age and sex) with a median (IQR) follow-up of 6.8 (1.7) years were analysed. No significant association was found in walking, moderate, and vigorous activities. In summed activity, only the 3rd quartile showed a statistically significant inverse association with PDAC risk (aHR 0.67, 95% CI 0.52-0.86, p<0.01). For IPAQ activity, the moderate and high activity groups showed borderline statistically significant associations with incident PDAC (aHR 0.80, 95% CI 0.63-1.00, p=0.05, and aHR 0.80, 95% CI 0.64-1.01, p=0.06, respectively). Conclusion The large UK Biobank cohort study did not show a strong association between physical activity levels and the development of incident PDAC.
Collapse
Affiliation(s)
- Borna A Assarian
- Human Development and Health, Faculty of Medicine University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, GBR
| | - Christopher D Byrne
- Human Development and Health, Faculty of Medicine University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, GBR
| | - Declan McDonnell
- Hepato-Pancreato-Biliary (HPB) Unit, University Hospital Southampton NHS Foundation Trust, Southampton, GBR
| | - Zaed Hamady
- Hepato-Pancreato-Biliary (HPB) Unit, University Hospital Southampton NHS Foundation Trust, Southampton, GBR
| |
Collapse
|
24
|
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
|
25
|
Belfrage H, Kuuliala K, Kuuliala A, Mustonen H, Puolakkainen P, Kylänpää L, Seppänen H, Louhimo J. Circulating necroptosis markers in chronic pancreatitis and pancreatic cancer: Associations with diagnosis and prognostic factors. Pancreatology 2024; 24:1229-1236. [PMID: 39613683 DOI: 10.1016/j.pan.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/01/2024]
Abstract
OBJECTIVES Necroptosis, a programmed inflammatory cell death, is implicated in the pathogenesis of pancreatitis and its potential progression to pancreatic ductal adenocarcinoma (PDAC), but its role remains unclear. We compared plasma levels of necroptosis-related markers - mixed lineage kinase domain-like protein (MLKL), interleukin (IL)-33, and its soluble receptor (sST2)- in patients with chronic pancreatitis (CP), PDAC, and healthy controls (HC), to explore their diagnostic and prognostic significance. METHODS Plasma was collected from patients pre-procedurally (PDAC, n = 82; CP, n = 25) and from HC (n = 39), and studied by ELISA. Clinical and routine laboratory data were collected, and pancreas was defined as soft or non-soft based on intraoperative or imaging findings. Mann-Whitney U test, Spearman correlation coefficients, ROC analysis were used for statistical analysis. RESULTS Plasma MLKL was lower in patients with CP than in other groups (p < 0.001) and PDAC patients treated with upfront surgery (PDACUS, n = 65) had lower MLKL than HC (p = 0.035). MLKL differentiated CP from PDACUS (AUC 0.83, p < 0.001). sST2 levels were significantly lower in HC than in other groups (p < 0.001) and in PDAC patients with a soft pancreas compared with non-soft (p < 0.005). In Lewis antigen positive PDACUS (n = 59), sST2 had positive correlations with CA19-9 measured concurrently and after 1 month, and with CEA measured after 1 or 6 months. CONCLUSIONS Circulating levels of MLKL are lower in patients with CP than PDAC. Elevated sST2 levels are associated with pancreatic diseases. Further studies are required to show whether MLKL and sST2 could be useful biomarkers in evaluating pancreatic diseases.
Collapse
Affiliation(s)
- Hanna Belfrage
- Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.
| | - Krista Kuuliala
- Department of Bacteriology and Immunology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Antti Kuuliala
- Department of Bacteriology and Immunology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Harri Mustonen
- Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Translational Cancer Medicine Program and ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Pauli Puolakkainen
- Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Translational Cancer Medicine Program and ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Leena Kylänpää
- Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Hanna Seppänen
- Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Translational Cancer Medicine Program and ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Johanna Louhimo
- Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
26
|
Takuma K, Okano N, Kimura Y, Hoshi K, Sato Y, Ujita W, Iwata S, Nakagawa H, Watanabe K, Yamada Y, Iwasaki S, Igarashi Y, Matsuda T. Development of irreversible pancreatic ductal change triggered by pancreatic duct stenting in chronic pancreatitis. J Gastroenterol Hepatol 2024; 39:2746-2751. [PMID: 39227734 DOI: 10.1111/jgh.16733] [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: 02/27/2024] [Revised: 08/09/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND AND AIM Stent-induced ductal change is a complication of endoscopic pancreatic stent placement for chronic pancreatitis, potentially leading to irreversible changes that may contribute to pancreatic dysfunction. This study aimed to examine the long-term outcomes of stent-induced ductal change and evaluate factors that correlate with the development of irreversible ductal changes. METHODS Between January 2008 and December 2022, 52/223 patients with chronic pancreatitis in whom an S-type plastic stent was successfully placed from the main papilla for duct stricture were detected with stent-induced ductal change on pancreatography at stent removal. We retrospectively investigated the clinical features of patients whose main pancreatic duct was reassessed by endoscopic pancreatography after >1 month without stent and whose residual stent-induced ductal change was irreversible. RESULTS The patients with chronic pancreatitis with stent-induced ductal change (n = 28) (elevated change, 15; stricture change, 13) were evaluated using follow-up pancreatography. Eleven patients (39.3%) showed residual change associated with stent-induced ductal change, the degree of which was partial improvement, no change, and obstructive change in one, seven, and three patients, respectively. Stricture changes during stent removal and duration of stent placement that triggered ductal changes were significantly associated with the development of residual ductal changes. CONCLUSIONS Irreversible stent-induced ductal change in patients with chronic pancreatitis was associated with stricture changes in the main pancreatic duct and continued plastic-stent placement. Careful evaluation of the pancreatic duct is required during plastic-stent placement. Early plastic-stent removal may result in an effective response to the development of stent-induced ductal change.
Collapse
Affiliation(s)
- Kensuke Takuma
- Division of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan
| | - Naoki Okano
- Division of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan
| | - Yusuke Kimura
- Division of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan
| | - Kensuke Hoshi
- Division of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan
| | - Yoichiro Sato
- Division of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan
| | - Wataru Ujita
- Division of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan
| | - Shuntaro Iwata
- Division of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan
| | - Hiroki Nakagawa
- Division of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan
| | - Koji Watanabe
- Division of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan
| | - Yuto Yamada
- Division of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan
| | - Susumu Iwasaki
- Division of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan
| | - Yoshinori Igarashi
- Division of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan
| | - Takahisa Matsuda
- Division of Gastroenterology and Hepatology, Toho University Omori Medical Center, Tokyo, Japan
| |
Collapse
|
27
|
Dai Y, Guan X, Guo F, Kong X, Ji S, Shang D, Bai C, Zhang Q, Zhao L. Botanical drugs and their natural compounds: a neglected treasury for inhibiting the carcinogenesis of pancreatic ductal adenocarcinoma. PHARMACEUTICAL BIOLOGY 2024; 62:853-873. [PMID: 39520705 PMCID: PMC11552278 DOI: 10.1080/13880209.2024.2421759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/25/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
CONTEXT Pancreatic ductal adenocarcinoma (PDAC), which is characterized by its malignant nature, presents challenges for early detection and is associated with a poor prognosis. Any strategy that can interfere with the beginning or earlier stage of PDAC greatly delays disease progression. In response to this intractable problem, the exploration of new drugs is critical to reduce the incidence of PDAC. OBJECTIVE In this study, we summarize the mechanisms of pancreatitis-induced PDAC and traditional Chinese medicine (TCM) theory and review the roles and mechanisms of botanical drugs and their natural compounds that can inhibit the process of pancreatitis-induced PDAC. METHODS With the keywords 'chronic pancreatitis', 'TCM', 'Chinese medicinal formulae', 'natural compounds', 'PDAC' and 'pancreatic cancer', we conducted an extensive literature search of the PubMed, Web of Science, and other databases to identify studies that effectively prevent PDAC in complex inflammatory microenvironments. RESULTS We summarized the mechanism of pancreatitis-induced PDAC. Persistent inflammatory microenvironments cause multiple changes in the pancreas itself, including tissue damage, abnormal cell differentiation, and even gene mutation. According to TCM, pancreatitis-induced PDAC is the process of 'dampness-heat obstructing the spleen and deficiency due to stagnation' induced by a variety of pathological factors. A variety of botanical drugs and their natural compounds, such as Chaihu classical formulae, flavonoids, phenolics, terpenoids, etc., may be potential drugs to interfere with the development of PDAC via reshaping the inflammatory microenvironment by improving tissue injury and pancreatic fibrosis. CONCLUSIONS Botanical drugs and their natural compounds show great potential for preventing PDAC in complex inflammatory microenvironments.
Collapse
Affiliation(s)
- Yunfei Dai
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xi Guan
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fangyue Guo
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xin Kong
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- College of pharmacy, Dalian Medical University, Dalian, China
| | - Shuqi Ji
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Dong Shang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Changchuan Bai
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qingkai Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Liang Zhao
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
28
|
Su N, Tang R, Zhang Y, Ni J, Huang Y, Liu C, Xiao Y, Zhu B, Zhao Y. Using machine learning to identify risk factors for pancreatic cancer: a retrospective cohort study of real-world data. Front Pharmacol 2024; 15:1510220. [PMID: 39640479 PMCID: PMC11617206 DOI: 10.3389/fphar.2024.1510220] [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: 10/12/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024] Open
Abstract
Objectives This study aimed to identify the risk factors for pancreatic cancer through machine learning. Methods We investigated the relationships between different risk factors and pancreatic cancer using a real-world retrospective cohort study conducted at West China Hospital of Sichuan University. Multivariable logistic regression, with pancreatic cancer as the outcome, was used to identify covariates associated with pancreatic cancer. The machine learning model extreme gradient boosting (XGBoost) was adopted as the final model for its high performance. Shapley additive explanations (SHAPs) were utilized to visualize the relationships between these potential risk factors and pancreatic cancer. Results The cohort included 1,982 patients. The median ages for pancreatic cancer and nonpancreatic cancer groups were 58.1 years (IQR: 51.3-64.4) and 57.5 years (IQR: 49.5-64.9), respectively. Multivariable logistic regression indicated that kirsten rats arcomaviral oncogene homolog (KRAS) gene mutation, hyperlipidaemia, pancreatitis, and pancreatic cysts are significantly correlated with an increased risk of pancreatic cancer. The five most highly ranked features in the XGBoost model were KRAS gene mutation status, age, alcohol consumption status, pancreatitis status, and hyperlipidaemia status. Conclusion Machine learning algorithms confirmed that KRAS gene mutation, hyperlipidaemia, and pancreatitis are potential risk factors for pancreatic cancer. Additionally, the coexistence of KRAS gene mutation and pancreatitis, as well as KRAS gene mutation and pancreatic cysts, is associated with an increased risk of pancreatic cancer. Our findings offered valuable implications for public health strategies targeting the prevention and early detection of pancreatic cancer.
Collapse
Affiliation(s)
- Na Su
- West China School of Pharmacy, Sichuan University, Chengdu, China
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Tang
- Institute of Medical Information, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Yice Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jiaqi Ni
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yimei Huang
- University of Florida Health Shands Hospital, Gainesville, FL, United States
| | - Chunqi Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuzhou Xiao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Baoting Zhu
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Vilà-Quintana L, Fort E, Pardo L, Albiol-Quer MT, Ortiz MR, Capdevila M, Feliu A, Bahí A, Llirós M, García-Velasco A, Morell Ginestà M, Laquente B, Pozas D, Moreno V, Garcia-Gil LJ, Duell EJ, Pimenoff VN, Carreras-Torres R, Aldeguer X. Metagenomic Study Reveals Phage-Bacterial Interactome Dynamics in Gut and Oral Microbiota in Pancreatic Diseases. Int J Mol Sci 2024; 25:10988. [PMID: 39456772 PMCID: PMC11507633 DOI: 10.3390/ijms252010988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Individuals with pancreatic-related health conditions usually show lower diversity and different composition of bacterial and viral species between the gut and oral microbiomes compared to healthy individuals. We performed a thorough microbiome analysis, using deep shotgun sequencing of stool and saliva samples obtained from patients with chronic pancreatitis (CP), pancreatic ductal adenocarcinoma (PDAC), and healthy controls (HCs).We observed similar microbiota composition at the species level in both the gut and oral samples in PDAC patients compared to HCs, among which the most distinctive finding was that the abundance of oral-originated Fusobacterium nucleatum species did not differ between the oral and the gut samples. Moreover, comparing PDAC patients with HCs, Klebsiella oxytoca was significantly more abundant in the stool samples of PDAC patients, while Streptococcus spp. showed higher abundance in both the oral and stool samples of PDAC patients. Finally, the most important finding was the distinctive gut phage-bacterial interactome pattern among PDAC patients. CrAssphages, particularly Blohavirus, showed mutual exclusion with K. oxytoca species, while Burzaovirus showed co-occurrence with Enterobacteriaceae spp., which have been shown to be capable of inducing DNA damage in human pancreatic cells ex vivo. The interactome findings warrant further mechanistic studies, as our findings may provide new insights into developing microbiota-based diagnostic and therapeutic methods for pancreatic diseases.
Collapse
Affiliation(s)
- Laura Vilà-Quintana
- Digestive Diseases and Microbiota Group, Girona Biomedical Research Institute (IDIBGI), 17190 Salt, Spain
| | - Esther Fort
- Department of Gastroenterology, Hospital Universitari de Girona Dr. Josep Trueta, 17007 Girona, Spain
| | - Laura Pardo
- Department of Gastroenterology, Hospital Universitari de Girona Dr. Josep Trueta, 17007 Girona, Spain
| | - Maria T. Albiol-Quer
- Hepato-Pancreato-Biliary Unit, Department of Surgery, Hospital Universitari de Girona Dr. Josep Trueta, 17007 Girona, Spain
| | - Maria Rosa Ortiz
- Department of Pathology, Hospital Universitari de Girona Dr. Josep Trueta, 17007 Girona, Spain
| | - Montserrat Capdevila
- Digestive Diseases and Microbiota Group, Girona Biomedical Research Institute (IDIBGI), 17190 Salt, Spain
| | - Anna Feliu
- Digestive Diseases and Microbiota Group, Girona Biomedical Research Institute (IDIBGI), 17190 Salt, Spain
| | - Anna Bahí
- Digestive Diseases and Microbiota Group, Girona Biomedical Research Institute (IDIBGI), 17190 Salt, Spain
| | - Marc Llirós
- Bioinformatics and Bioimaging (BI-SQUARED) Research Group, Biosciences Department, Faculty of Sciences, Technology and Engineering, Universitat de Vic, 08500 Vic, Spain
| | - Adelaida García-Velasco
- Precision Oncology Group, Girona Biomedical Research Institute (IDIBGI), 17190 Salt, Spain
- Institut Català d’Oncologia (ICO), Hospital Universitari de Girona Dr. Josep Trueta, 17007 Girona, Spain
| | - Mireia Morell Ginestà
- Hereditary Cancer Program, Institut Català d’Oncologia (ICO), Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), CIBERONC, 08908 Barcelona, Spain
| | - Berta Laquente
- Medical Oncology Department, Institut Català d’Oncologia (ICO), Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), 08908 Barcelona, Spain
| | - Débora Pozas
- Digestive Diseases and Microbiota Group, Girona Biomedical Research Institute (IDIBGI), 17190 Salt, Spain
| | - Victor Moreno
- Institut Català d’Oncologia (ICO), Institut de Recerca Biomedica de Bellvitge (IDIBELL), 08908 Barcelona, Spain
- UBICS, University of Barcelona (UB), 08028 Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 08036 Barcelona, Spain
| | - Librado Jesús Garcia-Gil
- Digestive Diseases and Microbiota Group, Girona Biomedical Research Institute (IDIBGI), 17190 Salt, Spain
| | - Eric Jeffrey Duell
- Cancer Epidemiology Research Program, Unit of Nutrition and Cancer, Institut Català d’Oncologia (ICO), Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), 08908 Barcelona, Spain
| | - Ville Nikolai Pimenoff
- Department of Clinical Science, Intervention and Technology—CLINTEC, Karolinska Institutet, 14152 Stockholm, Sweden
- Unit of Population Health, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Robert Carreras-Torres
- Digestive Diseases and Microbiota Group, Girona Biomedical Research Institute (IDIBGI), 17190 Salt, Spain
| | - Xavier Aldeguer
- Digestive Diseases and Microbiota Group, Girona Biomedical Research Institute (IDIBGI), 17190 Salt, Spain
| |
Collapse
|
30
|
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
|
31
|
Chan CH, Chang CC, Peng YC. The Clinical Significance of Pancreatic Steatosis in Pancreatic Cancer: A Hospital-Based Study. Diagnostics (Basel) 2024; 14:2128. [PMID: 39410531 PMCID: PMC11475449 DOI: 10.3390/diagnostics14192128] [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: 07/17/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Pancreatic cancer remains one of the deadliest malignancies worldwide with a pressing need for early detection and intervention strategies. Emerging evidence has suggested a potential link between pancreas steatosis, characterized by excessive pancreatic fat accumulation, and an increased risk of pancreatic cancer development. This retrospective imaging study aims to elucidate the association between pancreatic steatosis and the subsequent development of pancreatic cancer. In the study, we aimed to determine the characteristics of pancreatic cancer with pancreatic steatosis. Methods: During the period of January 2022 to December 2022, we conducted a retrospective study, collecting 101 newly diagnosed pancreas cancer cases from the available image datasets. A comprehensive database of retrospective abdominal imaging studies, comprising computed tomography (CT) and magnetic resonance imaging (MRI), was established from a diverse patient population and subsequently analyzed. Inclusion criteria encompassed patients having available baseline imaging data, allowing for the assessment of pancreatic fat content. Pancreatic fat content was quantified using validated radiological techniques, while demographic, clinical, and histopathological data were all collected. The clinical data and patient characteristics were collected from medical records and analyzed. Results: Preliminary analysis revealed a significant correlation between elevated pancreatic fat content and an increased incidence of subsequent pancreatic cancer. Moreover, subgroup analysis based on age, gender, and comorbidities provided valuable insight into potential risk factors associated with this progression. Additionally, the study identified novel radiological markers that may serve as early indicators of pancreatic cancer development in individuals with pancreatic steatosis. Conclusions: In the imaging study, approximately 30% (30/101) of pancreatic cancer patients presented with pancreatic steatosis. Chronic pancreatitis emerged as the primary factor contributing to pancreatic steatosis in these patients. Importantly, pancreatic steatosis did not significantly impact the prognosis of pancreatic cancer. Follow-up data revealed no significant differences in survival duration between patients with or without pancreatic steatosis. Additionally, no association was found between pancreatic steatosis and hepatic steatosis.
Collapse
Affiliation(s)
- Chia-Hao Chan
- Department of Radiology, Taipei Veterans General Hospital Taitung Branch, Taitung 950410, Taiwan;
- Department of Radiology, Taichung Veterans General Hospital, Taichung 407219, Taiwan
| | - Chia-Chen Chang
- Department of Medical Imaging, China Medical University Hospital, China Medical University, Taichung 404327, Taiwan;
| | - Yen-Chun Peng
- Division of Gastroenterology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
32
|
Qin A, Shi K, Tindall RR, Li J, Cheng B, Li J, Yang B, Yu Q, Zhang Y, Hong B, Kaur B, Younes M, Shen Q, Bailey-Lundberg JM, Cao Y, Ko TC. Characterization of Pancreatic Collagen-Expressing Fibroblasts in Mouse Acute Pancreatitis. GASTRO HEP ADVANCES 2024; 4:100557. [PMID: 39866719 PMCID: PMC11761323 DOI: 10.1016/j.gastha.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 09/16/2024] [Indexed: 01/28/2025]
Abstract
Background and Aims Pancreatic stellate cells (PSCs) are critical mediators in chronic pancreatitis with an undefined role in acute pancreatitis (AP). PSCs consist of a heterogenous group of cells and are considered interchangeable with pancreatic fibroblasts. This study explored the heterogeneous nature of PSCs by characterizing pancreatic collagen-expressing fibroblasts (PCFs) via lineage tracing in mouse normal and AP pancreas and determining the effect of PCF depletion in AP. Methods Tandem dimer Tomato (tdTom+) PCFs in collagen type 1 (Col1)a2CreERtdTomato (Tom) mice receiving tamoxifen were characterized via fluorescence, Oil Red staining, and flow cytometry. AP was induced by cerulein, AP injury was assessed, and tdTom+ PCFs were monitored. The effect of PCF depletion on AP injury was evaluated in Col1a2CreERdiphtheria toxin A mice. Results Approximately 13% of pancreatic cells in Col1a2CreERTom mice were labeled by tdTom (tdTom+ PCFs), which surrounded acini, ducts, and blood vessels, and stained with Oil Red, collagen type I, vimentin, and desmin. tdTom+ PCFs increased 2-fold during AP, correlating with AP score, amylase, and alpha-smooth muscle actin+ and Ki67+ staining. PCF depletion in Col1a2CreERdiphtheria toxin A mice receiving tamoxifen resulted in enhanced inflammation compared to control. Conclusion PCFs may constitute a subset of PSCs and can be activated during AP. PCF depletion aggravates AP, suggesting a protective role for PCFs.
Collapse
Affiliation(s)
- Amy Qin
- Department of Surgery, UTHealth at Houston, Houston, Texas
| | - Kevin Shi
- Department of Surgery, UTHealth at Houston, Houston, Texas
| | | | - Jiajing Li
- Department of Surgery, UTHealth at Houston, Houston, Texas
| | - Binglu Cheng
- Department of Surgery, UTHealth at Houston, Houston, Texas
| | - Jing Li
- Department of Surgery, UTHealth at Houston, Houston, Texas
| | - Baibing Yang
- Department of Surgery, UTHealth at Houston, Houston, Texas
| | - Qiang Yu
- Department of Surgery, UTHealth at Houston, Houston, Texas
| | - Yinjie Zhang
- Department of Surgery, UTHealth at Houston, Houston, Texas
| | - Bangxing Hong
- Department of Pathology, Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Balveen Kaur
- Department of Pathology, Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Mamoun Younes
- Department of Pathology, George Washington University, Washington, District of Columbia
| | - Qiang Shen
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | | | - Yanna Cao
- Department of Surgery, UTHealth at Houston, Houston, Texas
| | - Tien C. Ko
- Department of Surgery, UTHealth at Houston, Houston, Texas
| |
Collapse
|
33
|
Gheorghe G, Diaconu CC, Mambet C, Bleotu C, Ionescu VA, Diaconu CC. Comparative analysis of leptin and carcinoembryonic antigen-related cell adhesion molecule 1 plasma expression in pancreatic cancer and chronic pancreatitis patients. Heliyon 2024; 10:e37410. [PMID: 39296050 PMCID: PMC11408808 DOI: 10.1016/j.heliyon.2024.e37410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/21/2024] Open
Abstract
Compared to the general population, patients with chronic pancreatitis have an up to 12-fold higher risk of developing pancreatic cancer. The aim of our study was the identification of potential proteomic biomarkers to contribute to the detection of pancreatic cancer among patients with chronic pancreatitis. We initially performed a proteomic screening analysis of 105 analytes on plasma pools. To validate this finding, we quantitatively determined leptin concentrations in individual plasma samples using the ELISA technique. Additionally, we explored the plasma expression of CEACAM1, an important regulator of leptin expression in various cancer cells using the same method. The preliminary semi-quantitative proteomic analysis identified leptin as the only protein with substantially higher expression in patients with pancreatic cancer compared to those with chronic pancreatitis. Subsequently, by quantitative ELISA, we determined a higher median leptin concentration in the plasma of patients with pancreatic cancer compared to those with chronic pancreatitis. The statistical significance was maintained regardless of other variables like BMI or gender. Additionally, we explored the plasma expression of CEACAM1, an important regulator of leptin expression in various cancer cells, in order to provide insights into the complex mechanisms underlying pancreatic cancer and chronic pancreatits. CEACAM1 concentrations were higher in the plasma of the patients with pancreatic cancer than in those with chronic pancreatitis. However, we did not find a statistically significant correlation between leptin and CEACAM1 expression variation in the two study groups, with CEACAM1 concentration also dependent on other parameters such as BMI, gender, and serum triglyceride level. In conclusion, leptin seems to be a biomarker that can contribute to differentiate patients with pancreatic cancer from patients with chronic pancreatitis.
Collapse
Affiliation(s)
- Gina Gheorghe
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474, Bucharest, Romania
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402, Bucharest, Romania
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304, Bucharest, Romania
| | - Carmen Cristina Diaconu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304, Bucharest, Romania
| | - Cristina Mambet
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474, Bucharest, Romania
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304, Bucharest, Romania
| | - Coralia Bleotu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304, Bucharest, Romania
| | - Vlad Alexandru Ionescu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474, Bucharest, Romania
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402, Bucharest, Romania
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304, Bucharest, Romania
| | - Camelia Cristina Diaconu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474, Bucharest, Romania
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402, Bucharest, Romania
| |
Collapse
|
34
|
Jiang D, Wu Y, Liu L, Shen Y, Li T, Lu Y, Wang P, Sun C, Wang K, Wang K, Ye H. Burden of Gastrointestinal Tumors in Asian Countries, 1990-2021: An Analysis for the Global Burden of Disease Study 2021. Clin Epidemiol 2024; 16:587-601. [PMID: 39252850 PMCID: PMC11381218 DOI: 10.2147/clep.s472553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 08/15/2024] [Indexed: 09/11/2024] Open
Abstract
Background Gastrointestinal tumors represent a significant component of the cancer burden in Asia. This study aims to evaluate the burden of gastrointestinal tumors in Asia from 1990 to 2021 using data from the Global Burden of Disease Study 2021 (GBD 2021). Methods The absolute incidence, mortality, and disability adjusted life years (DALYs) number and rate of six gastrointestinal tumors(colon and rectum cancer (CRC), stomach cancer (SC), pancreatic cancer (PC), esophageal cancer (EC), liver cancer (LC) and gallbladder and biliary tract cancer (GBTC)) in 48 Asian countries were extracted from GBD 2021. Differences were analyzed based on gender, age, year, location and socio-demographic index (SDI). Results In 2021, SC accounted for the highest disease burden in Asia (DALYs=16.41million [95% UI: 13.70, 19.62]). From 1990 to 2021, the age-standardized incidence rates of EC, LC, and SC in Asia declined, while the incidence rates of CRC and PC increased significantly, with CRC showing the largest rise (AAPC=1.08 [95% CI: 1.02 to 1.12]). Gastrointestinal tumors DALY rates peaked at age 70 and above, with males generally exhibiting higher rates than females. Furthermore, East Asia bears a higher burden compared to other Asian subregions. A higher SDI correlates with increased DALY rates for PC, but no linear relationship was observed for other gastrointestinal tumors. Conclusion The burden of gastrointestinal tumors in Asia remains high and may continue to increase. Therefore, effective prevention and treatment measures are essential to address the challenge posed by gastrointestinal tumors.
Collapse
Affiliation(s)
- Donglin Jiang
- College of Public Health, Zhengzhou University, Gaoxin District, Zhengzhou, Henan, 450001, People's Republic of China
| | - Yangxue Wu
- College of Public Health, Zhengzhou University, Gaoxin District, Zhengzhou, Henan, 450001, People's Republic of China
| | - Ling Liu
- College of Public Health, Zhengzhou University, Gaoxin District, Zhengzhou, Henan, 450001, People's Republic of China
| | - Yajing Shen
- College of Public Health, Zhengzhou University, Gaoxin District, Zhengzhou, Henan, 450001, People's Republic of China
| | - Tiandong Li
- College of Public Health, Zhengzhou University, Gaoxin District, Zhengzhou, Henan, 450001, People's Republic of China
| | - Yin Lu
- College of Public Health, Zhengzhou University, Gaoxin District, Zhengzhou, Henan, 450001, People's Republic of China
| | - Peng Wang
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China
| | - Changqing Sun
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China
| | - Kaijuan Wang
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China
| | - Keyan Wang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China
| | - Hua Ye
- College of Public Health, Zhengzhou University, Gaoxin District, Zhengzhou, Henan, 450001, People's Republic of China
| |
Collapse
|
35
|
Brasky TM, Jager LR, Newton AM, Li X, Loomans-Kropp HA, Hays JL, Margolis KL, Luo J. Use of Nonsteroidal Anti-Inflammatory Drugs and Pancreatic Cancer Risk in the Women's Health Initiative. Cancer Epidemiol Biomarkers Prev 2024; 33:1203-1210. [PMID: 38900510 PMCID: PMC11371515 DOI: 10.1158/1055-9965.epi-24-0305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/03/2024] [Accepted: 06/14/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Pancreatic cancer is among the most fatal human cancers and the fourth leading cause of cancer death in the United States. Evidence suggests that chronic inflammation may play a role in pancreatic carcinogenesis and its inhibition through nonsteroidal anti-inflammatory drugs (NSAID) may reduce pancreatic cancer incidence. METHODS We examined associations of total and individual NSAIDs with pancreatic cancer risk among postmenopausal women participating in the Women's Health Initiative observational study and clinical trial cohorts. Among 117,452 women, aged 55 to 79 years, 727 incident pancreatic cancer cases were reported over 18 years of follow-up. Cox regression was used to estimate hazard ratio (HR) and 95% confidence interval (CI) for associations between NSAIDs and pancreatic cancer risk. RESULTS Relative to non-use, consistent use of any NSAID was inversely associated with pancreatic cancer risk (HR 0.71, 95% CI, 0.59-0.87), primarily driven by strong associations for aspirin use (HR 0.67, 95% CI, 0.52-0.86). Use of total or individual non-aspirin NSAIDs was not associated with pancreatic cancer. Upon stratified analysis, we observed stronger associations for NSAIDs among participants with prevalent diabetes (HR 0.28, 95% CI, 0.10-0.75) relative to those without (HR 0.75, 95% CI, 0.61-0.92; P-interaction = 0.03). CONCLUSIONS Additional large prospective studies with careful measurement of NSAID type, dose, and frequency are needed to further investigate the possibility of added benefit among individuals diagnosed with diabetes. IMPACT This study adds to existing evidence from prospective studies and clinical trials suggesting that use of aspirin may provide moderate benefit for pancreatic cancer prevention.
Collapse
Affiliation(s)
- Theodore M. Brasky
- Division of Medical Oncology, The Ohio State University College of Medicine; Columbus, OH, USA
- The Ohio State University – Comprehensive Cancer Center, Cancer Control Research Program; Columbus, OH, USA
| | - Leah R. Jager
- Division of Public Health Sciences, Fred Hutchinson Cancer Center; Seattle, WA, USA
| | - Alison M. Newton
- Division of Medical Oncology, The Ohio State University College of Medicine; Columbus, OH, USA
- The Ohio State University – Comprehensive Cancer Center, Cancer Control Research Program; Columbus, OH, USA
| | - Xilin Li
- Department of Environmental and Occupational Health, Indiana University School of Public Health, Bloomington, IN, USA
| | - Holli A. Loomans-Kropp
- The Ohio State University – Comprehensive Cancer Center, Cancer Control Research Program; Columbus, OH, USA
- Division of Cancer Prevention and Control, The Ohio State University College of Medicine; Columbus, OH, USA
| | - John L. Hays
- Division of Medical Oncology, The Ohio State University College of Medicine; Columbus, OH, USA
| | | | - Juhua Luo
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health, Bloomington, IN, USA
| |
Collapse
|
36
|
Hong J, Fu Y, Chen X, Zhang Y, Li X, Li T, Liu Y, Fan M, Lin R. Gut microbiome changes associated with chronic pancreatitis and pancreatic cancer: a systematic review and meta-analysis. Int J Surg 2024; 110:5781-5794. [PMID: 38847785 PMCID: PMC11392207 DOI: 10.1097/js9.0000000000001724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/19/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND The study of changes in the microbiome in chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC) holds significant potential for developing noninvasive diagnostic tools as well as innovative interventions to alter the progression of diseases. This systematic review and meta-analysis aimed to analyze in detail the taxonomic and functional characteristics of the gut microbiome in patients with CP and PDAC. METHODS Two researchers conducted a systematic search across public databases to gather all published research up to June 2023. Diversity and gut microbiota composition are the main outcomes the authors focus on. RESULTS This meta-analysis included 14 studies, involving a total of 1511 individuals in the PDAC ( n =285), CP ( n =342), and control ( n =649) groups. Our results show a significant difference in the composition of gut microbiota between PDAC/CP patients compared to healthy controls (HC), as evidenced by a slight decrease in α-diversity, including Shannon (SMD=-0.33; P =0.002 and SMD=-0.59; P <0.001, respectively) and a statistically significant β-diversity ( P <0.05). The pooled results showed that at the phylum level, the proportion of Firmicutes was lower in PDAC and CP patients than in HC patients. At the genus level, more than two studies demonstrated that four genera were significantly increased in PDAC patients compared to HC (e.g. Escherichia-Shigella and Veillonella ). CP patients had an increase in four genera (e.g. Escherichia-Shigella and Klebsiella ) and a decrease in eight genera (e.g. Coprococcus and Bifidobacterium ) compared to HC. Functional/metabolomics results from various studies also showed differences between PDAC/CP patients and HC. In addition, this study found no significant differences in gut microbiota between PDAC and CP patients. CONCLUSIONS Current evidence suggests changes in gut microbiota is associated with PDAC/CP, commonly reflected by a reduction in beneficial species and an increase in the pathogenic species. Further studies are needed to confirm these findings and explore therapeutic possibilities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mengke Fan
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
37
|
Chen M, Guo B, Cheng H, Wang W, Jin J, Zhang Y, Deng X, Yang W, Wu C, Gao X, Yu D, Feng W, Chen Y. Genetic Engineering Bacillus thuringiensis Enable Melanin Biosynthesis for Anti-Tumor and Anti-Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308506. [PMID: 38943265 PMCID: PMC11423088 DOI: 10.1002/advs.202308506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/19/2024] [Indexed: 07/01/2024]
Abstract
Collaboration between cancer treatment and inflammation management has emerged as an integral facet of comprehensive cancer care. Nevertheless, the development of interventions concurrently targeting both inflammation and cancer has encountered significant challenges stemming from various external factors. Herein, a bioactive agent synthesized by genetically engineering melanin-producing Bacillus thuringiensis (B. thuringiensis) bacteria, simultaneously achieves eco-friendly photothermal agent and efficient reactive oxygen/nitrogen species (RONS) scavenger benefits, perfectly tackling present toughies from inflammation to cancer therapies. The biologically derived melanin exhibits exceptional photothermal-conversion performance, facilitating potent photonic hyperthermia that effectively eradicates tumor cells and tissues, thereby impeding tumor growth. Additionally, the RONS-scavenging properties of melanin produced by B. thuringiensis bacteria contribute to inflammation reduction, augmenting the efficacy of photothermal tumor repression. This study presents a representative paradigm of genetic engineering in B. thuringiensis bacteria to produce functional agents tailored for diverse biomedical applications, encompassing inflammation and cancer therapy.
Collapse
Affiliation(s)
- Meng Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Bingbing Guo
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Hui Cheng
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Weiyi Wang
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Junyi Jin
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Yingyi Zhang
- School of MedicineShenzhen Campus of SunYat‐Sen UniversityShenzhen518107P. R. China
- Center for Materials Synthetic BiologyCAS Key Laboratory of Quantitative Engineering BiologyShenzhen Institute of Synthetic BiologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518000P. R. China
| | - Xiaolian Deng
- School of MedicineShenzhen Campus of SunYat‐Sen UniversityShenzhen518107P. R. China
- Center for Materials Synthetic BiologyCAS Key Laboratory of Quantitative Engineering BiologyShenzhen Institute of Synthetic BiologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518000P. R. China
| | - Wenjun Yang
- Center for Materials Synthetic BiologyCAS Key Laboratory of Quantitative Engineering BiologyShenzhen Institute of Synthetic BiologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518000P. R. China
| | - Chenyao Wu
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Xiang Gao
- Center for Materials Synthetic BiologyCAS Key Laboratory of Quantitative Engineering BiologyShenzhen Institute of Synthetic BiologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518000P. R. China
| | - Dehong Yu
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Wei Feng
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
- School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative MedicineVision and Brain Health) Wenzhou Institute of Shanghai UniversityWenzhouZhejiang325088P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
- School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative MedicineVision and Brain Health) Wenzhou Institute of Shanghai UniversityWenzhouZhejiang325088P. R. China
| |
Collapse
|
38
|
Shah Y, Dahiya DS, Tiwari A, Kumar H, Gangwani MK, Ali H, Hayat U, Alsakarneh S, Singh S, Malik S, Sohail AH, Chandan S, Ali MA, Inamdar S. Advancements in Early Detection and Screening Strategies for Pancreatic Cancer: From Genetic Susceptibility to Novel Biomarkers. J Clin Med 2024; 13:4706. [PMID: 39200847 PMCID: PMC11355237 DOI: 10.3390/jcm13164706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
Pancreatic cancer is a rare but lethal cancer due to its biologically aggressive nature, advanced stage at the time of diagnosis, and poor response to oncologic therapies. The risk of pancreatic cancer is significantly higher to 5% in certain high-risk individuals with inherited genetic susceptibility. Screening for pancreatic cancer in these individuals from high-risk groups can help with the early detection of pancreatic cancer as well as the detection of precursor lesions leading to early surgical resection and improved overall outcomes. The advancements in radiological imaging as well as advanced endoscopic procedures has made a significant impact on the early diagnosis, surveillance, and staging of pancreatic cancer. There is also a significant advancement in the development of biomarkers for the early detection of pancreatic cancer, which has also led to the development of liquid biopsy, allowing for microRNA detection in serum and circulating tumor cells. Various societies and organizations have provided guidelines for pancreatic cancer screening and surveillance in high-risk individuals. In this review, we aim to discuss the hereditary risk factors for developing pancreatic cancer, summarize the screening recommendations by different societies, and discuss the development of novel biomarkers and areas for future research in pancreatic cancer screening for high-risk individuals.
Collapse
Affiliation(s)
- Yash Shah
- Department of Internal Medicine, Trinity Health Oakland/Wayne State University, Pontiac, MI 48341, USA
| | - Dushyant Singh Dahiya
- Division of Gastroenterology, Hepatology & Motility, The University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | - Angad Tiwari
- Department of Internal Medicine, Maharani Laxmi Bai Medical College, Jhansi 284001, Uttar Pradesh, India
| | - Harendra Kumar
- Department of Internal Medicine, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Manesh Kumar Gangwani
- Department of Gastroenterology and Hepatology, University of Arkansas For Medical Sciences, Little Rock, AR 72205, USA
| | - Hassam Ali
- Division of Gastroenterology, Hepatology & Nutrition, East Carolina University/Brody School of Medicine, Greenville, NC 27834, USA
| | - Umar Hayat
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes Barre, PA 18711, USA
| | - Saqr Alsakarneh
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Sahib Singh
- Department of Internal Medicine, Sinai Hospital, Baltimore, MD 21215, USA
| | - Sheza Malik
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY 14621, USA
| | - Amir H. Sohail
- Department of Surgery, University of New Mexico, Albuquerque, NM 87131, USA
| | - Saurabh Chandan
- Center for Interventional Endoscopy (CIE), Advent Health, Orlando, FL 32803, USA
| | - Meer A. Ali
- Department of Gastroenterology and Hepatology, University of Arkansas For Medical Sciences, Little Rock, AR 72205, USA
| | - Sumant Inamdar
- Department of Gastroenterology and Hepatology, University of Arkansas For Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
39
|
Treekitkarnmongkol W, Dai J, Liu S, Sankaran D, Nguyen T, Balasenthil S, Hurd MW, Chen M, Katayama H, Roy-Chowdhuri S, Calin GA, Brand RE, Lampe PD, Hu TY, Maitra A, Koay EJ, Killary AM, Sen S. Blood-Based microRNA Biomarker Signature of Early-Stage Pancreatic Ductal Adenocarcinoma With Lead-Time Trajectory in Prediagnostic Samples. GASTRO HEP ADVANCES 2024; 3:1098-1115. [PMID: 39529638 PMCID: PMC11550741 DOI: 10.1016/j.gastha.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/02/2024] [Indexed: 11/16/2024]
Abstract
Background and Aims Clinically validated biomarker of pancreatic ductal adenocarcinoma (PDAC), carbohydrate antigen 19-9 (CA19-9), has limited sensitivity and specificity for early-stage disease. Circulating miRNAs in plasma associated with cancer relevant pathways were developed as early detection biomarkers. Methods 2083 miRNAs in 15 μl of plasma from multicenter age-matched cohorts (N = 203: healthy controls, n = 46; pancreatitis controls, n = 36; diagnosed cases: n = 121) and a prediagnostic Prostate, Lung, Colorectal, and Ovarian age- and gender-matched cohort (N = 96; controls, n = 48; prediagnosed cases, n = 48) were interrogated. A three-miRNA biomarker signature was developed for early-stage PDAC. Results The three-miRNA signature (let-7i-5p, miR-130a-3p and miR-221-3p) detected PDAC from healthy controls independently (area under the curve [AUC] of stage I, II, I-IV = 0.970, 0.975, 0.974) and in combination with CA19-9 (AUC of stage I, II, I-IV = 1.000, 0.992, 0.995). It also discriminated chronic pancreatitis (AUC of stage I, II, I-IV = 0.932, 0.931, 0.929), improving performance of CA19-9 alone (AUC of stage I, II, I-IV = 0.763, 0.701, 0.735) in combination (AUC of stage I, II, I-IV = 0.971, 0.943, 0.951). Blinded validation in prediagnostic Prostate, Lung, Colorectal, and Ovarian cohort revealed lead-time trajectory increase in AUC from 0.702 to 0.729 to 0.757 at twelve-, six-, and three-months before PDAC diagnosis, respectively. The signature also helped stratification of patients with different circulating tumor DNA and imaging subtypes. Conclusion Plasma miRNAs associated with oncogenic pathways may serve as PDAC early detection biomarkers.
Collapse
Affiliation(s)
- Warapen Treekitkarnmongkol
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jianliang Dai
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Suyu Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Deivendran Sankaran
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tristian Nguyen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Seetharaman Balasenthil
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark W. Hurd
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Meng Chen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hiroshi Katayama
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sinchita Roy-Chowdhuri
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - George A. Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Randall E. Brand
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Paul D. Lampe
- Translation Research Program, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Tony Y. Hu
- Department of Molecular & Cellular Biology, Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana
| | - Anirban Maitra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eugene J. Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ann M. Killary
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Subrata Sen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
40
|
Grigorescu RR, Husar-Sburlan IA, Gheorghe C. Pancreatic Cancer: A Review of Risk Factors. Life (Basel) 2024; 14:980. [PMID: 39202722 PMCID: PMC11355429 DOI: 10.3390/life14080980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Pancreatic adenocarcinoma is one of the most lethal types of gastrointestinal cancer despite the latest medical advances. Its incidence has continuously increased in recent years in developed countries. The location of the pancreas can result in the initial symptoms of neoplasia being overlooked, which can lead to a delayed diagnosis and a subsequent reduction in the spectrum of available therapeutic options. The role of modifiable risk factors in pancreatic cancer has been extensively studied in recent years, with smoking and alcohol consumption identified as key contributors. However, the few screening programs that have been developed focus exclusively on genetic factors, without considering the potential impact of modifiable factors on disease occurrence. Thus, fully understanding and detecting the risk factors for pancreatic cancer represents an important step in the prevention and early diagnosis of this type of neoplasia. This review reports the available evidence on different risk factors and identifies the areas that could benefit the most from additional studies.
Collapse
Affiliation(s)
- Raluca Roxana Grigorescu
- Gastroenterology Department, “Sfanta Maria” Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | | | - Cristian Gheorghe
- Center for Digestive Disease and Liver Transplantation, Fundeni Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
41
|
Satoh T, Nakatani E, Ariyasu H, Kawaguchi S, Ohno K, Itoh H, Hayashi K, Usui T. Pancreatic cancer risk in diabetic patients using the Japanese Regional Insurance Claims. Sci Rep 2024; 14:16958. [PMID: 39043788 PMCID: PMC11266625 DOI: 10.1038/s41598-024-67505-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 07/11/2024] [Indexed: 07/25/2024] Open
Abstract
Pancreatic cancer presents a critical health issue characterized by low survival rates. Identifying risk factors in specific populations, such as those with diabetes, is crucial for early detection and improved outcomes. This study aimed to identify risk factors for pancreatic cancer in diabetic patients using a longitudinal cohort from the Shizuoka Kokuho database, spanning April 2012 to September 2021. Diabetic patients were identified and monitored for the onset of pancreatic cancer. Factors analyzed included age, sex, the Elixhauser comorbidity index, and specific comorbidities. Statistical analyses involved univariate and multivariate Cox proportional hazards regression. The study identified 212,775 as diabetic patients and 1755 developed pancreatic cancer during the period. The annual incidence rate of pancreatic cancer in this group was 166.7 cases per 100,000 person-years. The study identified older age, male sex, a history of liver disease, chronic pancreatitis, and pancreatic cystic lesions as significant risk factors for pancreatic cancer in diabetic patients. The study also highlighted the absence of a significant association between diabetes type or diabetic complications and the onset of pancreatic cancer. These findings may aid in the early diagnosis of pancreatic cancer in diabetic patients and may inform revisions in screening practices in diabetic patients.
Collapse
Affiliation(s)
- Tatsunori Satoh
- Division of Endocrinology, Metabolism and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Shizuoka General Hospital, Shizuoka, Japan
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, 4-27-2 Kitaando, Aoi-Ku, Shizuoka, 420-0881, Japan
| | - Eiji Nakatani
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, 4-27-2 Kitaando, Aoi-Ku, Shizuoka, 420-0881, Japan.
| | - Hiroyuki Ariyasu
- Department of Diabetes and Endocrinology, Shizuoka General Hospital, Shizuoka, Japan
| | - Shinya Kawaguchi
- Department of Gastroenterology, Shizuoka General Hospital, Shizuoka, Japan
| | - Kazuya Ohno
- Department of Gastroenterology, Shizuoka General Hospital, Shizuoka, Japan
| | - Hiroshi Itoh
- Center for Preventive Medicine, Keio University, Tokyo, Japan
| | - Kaori Hayashi
- Division of Endocrinology, Metabolism and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takeshi Usui
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, 4-27-2 Kitaando, Aoi-Ku, Shizuoka, 420-0881, Japan
- Research Support Center, Shizuoka General Hospital, Shizuoka, Japan
| |
Collapse
|
42
|
Zhao JF, Zhou BG, Lv Y, Teng QP, Wang XM, Li XY, Ding Y. Association between metabolic dysfunction-associated steatotic liver disease and risk of colorectal cancer or colorectal adenoma: an updated meta-analysis of cohort studies. Front Oncol 2024; 14:1368965. [PMID: 39045565 PMCID: PMC11263091 DOI: 10.3389/fonc.2024.1368965] [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: 01/11/2024] [Accepted: 06/25/2024] [Indexed: 07/25/2024] Open
Abstract
Background and aims In recent years, the relationship between metabolic dysfunction-associated steatotic liver disease (MASLD) and colorectal cancer (CRC) or colorectal adenoma (CRA) has gained widespread attention. Previous meta-analyses on this subject either incorporated numerous cross-sectional studies, which were susceptible to bias, or concentrated solely on a restricted number of cohort studies. Moreover, with the release of a substantial number of high-quality cohort studies on this subject in the past two years, the findings continue to be debated and contradictory. Therefore, we conducted an updated systematic review and meta-analysis of cohort studies to quantitatively evaluate the magnitude of the association between them. Methods Comprehensive searches of PubMed, Web of Science, and Embase were conducted without language restrictions from the time of their creation up to December, 2023. The pooled hazard ratios (HRs) with 95% confidence interval (CIs) were calculated by the generic inverse variance based on the random-effects model. Moreover, subgroup and sensitivity analyses were performed. Results A total of 15 cohort studies were analyzed in this meta-analysis, which included 9,958,412 participants. The meta-analysis of 13 cohort studies showed that MASLD was linked to a higher risk of CRC (HR=1.25, 95% CI: 1.15-1.36, P < 0.00001). Additionally, further subgroup analysis indicated that the combined HR remained consistent regardless of the study location, nomenclature of fatty liver disease (FLD), confirmation methods for FLD, sample size, follow-up time, and study quality. Furthermore, the meta-analysis of four cohort studies demonstrated that MASLD was correlated with an increased risk of CRA (HR=1.38, 95% CI: 1.17-1.64, P = 0.0002). The sensitivity analysis results further validated the robustness of the aboved findings. Conclusion The results of our meta-analysis indicated that MASLD was associated with an increased risk of incident CRC/CRA. In the future, it is necessary to conduct more prospective cohort studies to thoroughly assess potential confounding factors, particularly in individuals from Europe and North America. Furthermore, related mechanism studies should be conducted to enhance our understanding of the link between MASLD and CRC/CRA. Systematic review registration Open Science Framework registries (https://osf.io/m3p9k).
Collapse
Affiliation(s)
- Jian-Feng Zhao
- Department of Gastrointestinal Surgery, Jingmen People’s Hospital, Jingchu University of Technology Affiliated Central Hospital, Jingmen, Hubei, China
| | | | - Yang Lv
- Department of Gastrointestinal Surgery, Jingmen People’s Hospital, Jingchu University of Technology Affiliated Central Hospital, Jingmen, Hubei, China
| | - Qiu-Ping Teng
- Department of Nephrology, The Central Hospital of Jingmen, Jingmen, Hubei, China
| | - Xi-Mei Wang
- Department of Gastrointestinal Surgery, Jingmen People’s Hospital, Jingchu University of Technology Affiliated Central Hospital, Jingmen, Hubei, China
| | - Xiao-Yi Li
- Imaging Diagnosis Center, Jingmen People’s Hospital, Jingchu University of Technology Affiliated Central Hospital, Jingmen, Hubei, China
| | - Yi Ding
- Department of Gastrointestinal Surgery, Jingmen People’s Hospital, Jingchu University of Technology Affiliated Central Hospital, Jingmen, Hubei, China
| |
Collapse
|
43
|
An H, Dai H, Liu X. Changing Trends in the Global Disease Burden of Pancreatic Cancer from 1990 to 2030. Dig Dis Sci 2024; 69:2450-2461. [PMID: 38722410 DOI: 10.1007/s10620-024-08465-y] [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: 03/03/2024] [Accepted: 04/23/2024] [Indexed: 07/19/2024]
Abstract
AIM To explore the global burden of pancreatic cancer (PC) from 1990 to 2019, evaluate independent effects of age, period, and cohort on the incidence of PC, and predict the incidence of PC in the next decade. METHODS Data were obtained from the Global Burden of Disease Study 2019. We calculated the age-standardized disability-adjusted life years (DALY) rate, age-standardized mortality rate (ASMR), age-standardized incidence rate (ASIR), and age-standardized prevalence rate (ASPR) of PC. Joinpoint Poisson regression analysis was performed to identify the temporal trends in the incidence of PC. Then, a two-factor model was constructed using the Poisson log-linear model, and a three-factor model was constructed using the intrinsic estimator (IE) method to estimate the independent effects of age, period, and cohort on the incidence of PC. Finally, the Bayesian age-period-cohort (BAPC) model was also used to predict the age-standardized global incidence rate of PC and age-standardized new PC cases from 2020 to 2030. RESULTS Overall, the DALY rate, ASMR, ASIR, and ASPR all increased from 1990 to 2019. The ASIR in males increased from 6 per 100,000 in 1990 to 7.5 per 100,000 in 2019 and was predicted to rise to 8.2 per 100,000 by 2030. Meanwhile, the ASIR in females rose from 4.5 per 100,000 in 1990 to 5.7 per 100,000 in 2019 and was predicted to rise to 6.3 per 100,000 by 2030. The age effect on the incidence of PC showed sharp increasing trends from 40 to 79 years. The period effect continuously increased with advancing periods, but the cohort effect showed substantial decreasing trends. CONCLUSIONS The age and period effect on the incidence of PC presented increasing trends, while the cohort effect showed decreasing trends. All indicators of the global burden of PC are increasing in both males and females, and the ASIR is predicted to rise at an alarming rate by 2030. Thus, timely screening and intervention are recommended, especially for earlier birth cohorts at high risk.
Collapse
Affiliation(s)
- Haoyu An
- School of Medicine, The Chinese University of Hong Kong, Shatin, 999077, NT, Hong Kong.
| | - Hanqian Dai
- Tianyuan Honors School, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Xiaomeng Liu
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
44
|
Danpanichkul P, Uawithya E, Lopimpisuth C, Sukphutanan B, Kulthamrongsri N, Aboona MB, Duangsonk K, Lau S, Simadibrata DM, Daggag H, Wallace MB, Wijarnpreecha K. Early-onset pancreatic cancer and associated metabolic risk factors in the Middle East and North Africa: A 20-year analysis of the Global Burden of Disease Study. Indian J Gastroenterol 2024:10.1007/s12664-024-01626-x. [PMID: 38951365 DOI: 10.1007/s12664-024-01626-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 05/30/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND AND OBJECTIVE Early-onset pancreatic cancer (EOPC) is associated with poor prognosis and high disease burden. Metabolic risk factors such as diabetes and obesity are considered risk factors of EOPC. Recently, there has been an increasing number of EOPCs worldwide. However, the analysis of EOPC, including its metabolic risk factors, in the Middle East and North Africa (MENA) region has not been fully addressed. METHODS Data from the Global Burden of Disease Study between 2000 and 2019 was used to analyze the prevalence, incidence, deaths and disability-adjusted life years (DALYs) associated with EOPC and its metabolic risk factors. The analysis further categorized the data based on countries, income status and sex and examined the annual percentage change (APC). RESULTS Approximately 2800 cases, 2400 deaths and 114,000 DALYs were attributable to EOPC in the MENA region. The incidence (APC + 3.42%), death (APC + 0.73%) and DALYs (APC + 3.23%) rates of EOPC increased. In addition, the death and DALY rates of EOPC attributable to obesity and diabetes increased. High and upper-middle-income countries exhibited a higher burden of EOPC than lower-income countries. CONCLUSION Over the past two decades, the burden of EOPC and its associated metabolic risk factors has increased. There is an urgent need for region-wide policy development, including screening methods and risk factor reduction, to mitigate the high and rising burden of EOPC in the MENA region.
Collapse
Affiliation(s)
- Pojsakorn Danpanichkul
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Immunology Unit, Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Ekdanai Uawithya
- Faculty of Medicine Siriraj Hospital, Mahidol University, Nakhon Pathom, Thailand
| | - Chawin Lopimpisuth
- Department of Internal Medicine, University of Miami, Jackson Memorial Hospital, Miami, FL, USA
| | | | - Narathorn Kulthamrongsri
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Nakhon Pathom, Thailand
- Department of Cardiovascular Medicine, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Majd B Aboona
- Department of Internal Medicine, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Kwanjit Duangsonk
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sirimas Lau
- Department of Internal Medicine, Metrowest Medical Center, Framingham, MA, USA
| | - Daniel M Simadibrata
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Hinda Daggag
- Imperial College London Diabetes Centre, Abu Dhabi, United Arab Emirates
| | - Michael B Wallace
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
- Department of Gastroenterology, Sheikh Shakhbout Medical City, Abu Dhabi, United Arab Emirates
| | - Karn Wijarnpreecha
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ, USA
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Banner University Medical Center, Phoenix, AZ, USA
- BIO5 Institute, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| |
Collapse
|
45
|
Joris S, Giron P, Olsen C, Seneca S, Gheldof A, Staessens S, Shahi RB, De Brakeleer S, Teugels E, De Grève J, Hes FJ. Identification of RAD17 as a candidate cancer predisposition gene in families with histories of pancreatic and breast cancers. BMC Cancer 2024; 24:723. [PMID: 38872153 PMCID: PMC11170902 DOI: 10.1186/s12885-024-12442-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 05/28/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Among the 10% of pancreatic cancers that occur in a familial context, around a third carry a pathogenic variant in a cancer predisposition gene. Genetic studies of pancreatic cancer predisposition are limited by high mortality rates amongst index patients and other affected family members. The genetic risk for pancreatic cancer is often shared with breast cancer susceptibility genes, most notably BRCA2, PALB2, ATM and BRCA1. Therefore, we hypothesized that additional shared genetic etiologies might be uncovered by studying families presenting with both breast and pancreatic cancer. METHODS Focusing on a multigene panel of 276 DNA Damage Repair (DDR) genes, we performed next-generation sequencing in a cohort of 41 families with at least three breast cancer cases and one pancreatic cancer. When the index patient with pancreatic cancer was deceased, close relatives (first or second-degree) affected with breast cancer were tested (39 families). RESULTS We identified 27 variants of uncertain significance in DDR genes. A splice site variant (c.1605 + 2T > A) in the RAD17 gene stood out, as a likely loss of function variant. RAD17 is a checkpoint protein that recruits the MRN (MRE11-RAD50-NBS1) complex to initiate DNA signaling, leading to DNA double-strand break repair. CONCLUSION Within families with breast and pancreatic cancer, we identified RAD17 as a novel candidate predisposition gene. Further genetic studies are warranted to better understand the potential pathogenic effect of RAD17 variants and in other DDR genes.
Collapse
Affiliation(s)
- Sofie Joris
- Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, Brussels, 1090, Belgium.
- The Oncology Research Center, the Laboratory for Medical & Molecular Oncology (LMMO), Faculty of Medicine, Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| | - Philippe Giron
- Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, Brussels, 1090, Belgium
| | - Catharina Olsen
- Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, Brussels, 1090, Belgium
| | - Sara Seneca
- Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, Brussels, 1090, Belgium
| | - Alexander Gheldof
- Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, Brussels, 1090, Belgium
| | - Shula Staessens
- The Oncology Research Center, the Laboratory for Medical & Molecular Oncology (LMMO), Faculty of Medicine, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Rajendra Bahadur Shahi
- The Oncology Research Center, the Laboratory for Medical & Molecular Oncology (LMMO), Faculty of Medicine, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Sylvia De Brakeleer
- The Oncology Research Center, the Laboratory for Medical & Molecular Oncology (LMMO), Faculty of Medicine, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Erik Teugels
- The Oncology Research Center, the Laboratory for Medical & Molecular Oncology (LMMO), Faculty of Medicine, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Jacques De Grève
- Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, Brussels, 1090, Belgium
- The Oncology Research Center, the Laboratory for Medical & Molecular Oncology (LMMO), Faculty of Medicine, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Frederik J Hes
- Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, Brussels, 1090, Belgium
| |
Collapse
|
46
|
Chang C, Yan HM, Liao YL. No association between hepatitis C virus infection and risk of colorectal cancer: a systematic review and meta-analysis of cohort studies. Front Med (Lausanne) 2024; 11:1327809. [PMID: 38898936 PMCID: PMC11186414 DOI: 10.3389/fmed.2024.1327809] [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: 10/25/2023] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Background and aim There is still uncertainty regarding whether hepatitis C virus (HCV) infection is associated with colorectal cancer (CRC). This study aims to investigate the potential association between HCV infection and CRC through a systematic review and meta-analysis of cohort studies. Methods PubMed, Embase, and Web of Science were systematically searched from the beginning of their inception to October 2023 to find relevant cohort studies on the association between HCV infection and CRC risk. The random-effect, generic inverse variance method was used to calculate the hazard ratios (HRs) and 95% confidence intervals (CIs) for CRC outcome among individuals with HCV infection. We also performed subgroup and sensitivity analysis. Results A total of 8 cohort studies involving 1,939,164 participants were included in this meta-analysis. The result from the meta-analysis suggested that there was no statistically significant association between HCV and the risk of developing CRC (HR = 0.99, 95% CI: 0.82-1.88, p = 0.88) with low statistical heterogeneity (I2 = 28%, p = 0.20). Subgroup analyses that were conducted based on study design, diagnosis of HCV infection, and publication year yielded similar results. Analyses of subgroups based on study areas revealed that there was no significant association between HCV infection and CRC risk in Asia (n = 2, HR = 0.96, 95% CI: 0.71-1.29, p = 0.79; I2 = 26%), Europe (n = 3, HR = 1.06, 95% CI: 0.83-1.37, p = 0.63; I2 = 0%), and North America (n = 2, HR = 1.10, 95% CI: 0.87-1.38, p = 0.44; I2 = 0%); however, a negative correlation was found in Oceania (n = 1, HR = 0.43, 95% CI: 0.22-0.84, p = 0.01). Sensitivity analysis further reinforce the stability of our conclusion. Conclusion Our cohort-based meta-analysis showed insufficient evidence to support the association between HCV infection and an increased risk of CRC. To gain a clearer insight into the potential association between these two conditions, it would be beneficial to conduct large, well-designed, high-quality prospective cohort studies that consider different ethnic populations and potential confounding factors.Systematic review registration: PROSPERO, identifier [CRD42023472688], https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023472688.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Gastroenterology, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Hong-Mei Yan
- Department of Gastroenterology, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Yan-Lin Liao
- Department of Cardiovascular Medicine, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
47
|
Sapoor S, Nageh M, Shalma NM, Sharaf R, Haroun N, Salama E, Pratama Umar T, Sharma S, Sayad R. Bidirectional relationship between pancreatic cancer and diabetes mellitus: a comprehensive literature review. Ann Med Surg (Lond) 2024; 86:3522-3529. [PMID: 38846873 PMCID: PMC11152885 DOI: 10.1097/ms9.0000000000002036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/30/2024] [Indexed: 06/09/2024] Open
Abstract
Pancreatic cancer (PC) is a fatal malignant disease. It is well known that the relationship between PC and type 2 diabetes mellitus (T2DM) is a complicated bidirectional relationship. The most important factors causing increased risks of pancreatic cancer are hyperglycaemia, hyperinsulinemia, pancreatitis, and dyslipidemia. Genetics and the immune system also play an important role in the relationship between diabetes mellitus and pancreatic cancer. The primary contributors to this association involve insulin resistance and inflammatory processes within the tumour microenvironment. The combination of diabetes and obesity can contribute to PC by inducing hyperinsulinemia and influencing leptin and adiponectin levels. Given the heightened incidence of pancreatic cancer in diabetes patients compared to the general population, early screening for pancreatic cancer is recommended. Diabetes negatively impacts the survival of pancreatic cancer patients. Among patients receiving chemotherapy, it reduced their survival. The implementation of a healthy lifestyle, including weight management, serves as an initial preventive measure to mitigate the risk of disease development. The role of anti-diabetic drugs on survival is controversial; however, metformin may have a positive impact, especially in the early stages of cancer, while insulin therapy increases the risk of PC.
Collapse
Affiliation(s)
| | | | | | - Rana Sharaf
- Faculty of Medicine, Alexandria University, Alexandria
| | - Nooran Haroun
- Faculty of Medicine, Alexandria University, Alexandria
| | - Esraa Salama
- Faculty of Medicine, Alexandria University, Alexandria
| | | | | | - Reem Sayad
- Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
48
|
Molero X, Ayuso JR, Balsells J, Boadas J, Busquets J, Casteràs A, Concepción M, Cuatrecasas M, Fernàndez Esparrach G, Fort E, Garcia Borobia F, Ginès À, Ilzarbe L, Loras C, Masachs M, Merino X, Olsina JJ, Puig-Diví V, Salord S, Serrano T, Vaquero EC. Chronic pancreatitis for the clinician: complications and special forms of the disease. Interdisciplinary position paper of the Catalan Society of Digestology (SCD) and the Catalan Pancreatic Society (SCPanc). Minerva Gastroenterol (Torino) 2024; 70:208-224. [PMID: 35262306 DOI: 10.23736/s2724-5985.22.03127-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chronic pancreatitis tends to develop a number of complications that may constitute the form of presentation of the disease. Some societies have issued guidelines for diagnosis and treatment of chronic pancreatitis complications, but the level of evidence for any topic is usually low and recommendations tend to be weak. We aimed to provide defined position statements for the clinician based on updated review of published literature and on multidisciplinary expert agreement. The goal was to propose defined terminology and rational diagnostic/therapeutic circuits based on current knowledge. To this end 14 sections related to complications and special forms of chronic pancreatitis (early chronic, groove and autoimmune pancreatitis) were reviewed by 21 specialists from 6 different fields to generate 32 statements. Featured statements assert common bile duct stenosis does not require invasive treatment (endoscopic or surgical) unless cholestasis, cholangitis, lithiasis or other symptoms develop. Pancreatic duct strictures and calculi should be approached (after ruling out malignancy) if causing pain, pancreatitis, pseudocysts or other complications. Treatment of symptomatic pseudocysts must be individualized, considering associated main duct stenosis, vascular and pericystic complications. Higher risk conditions for pancreatic cancer are advance age, smoking, genetic background, recent diagnosis of chronic pancreatitis or diabetes, and appearance of new symptoms. Groove pancreatitis can initially be treated with conservative measures. Both prednisolone or rituximab can induce remission and maintenance of autoimmune pancreatitis. Internal fistula, vascular complications, bacterial overgrowth, osteoporosis and renal lithiasis require specific therapeutic approaches.
Collapse
Affiliation(s)
- Xavier Molero
- Unit of Exocrine Pancreas Research, Department of Gastroenterology, VHIR, CIBERehd, Vall d'Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain -
| | - Juan R Ayuso
- Department of Radiology, CDI, IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Joaquim Balsells
- Department of Hepato-Pancreato-Biliary and Transplantation Surgery, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Jaume Boadas
- Department of Gastroenterology, Consorci Sanitari de Terrassa, Terrassa, Spain
| | - Juli Busquets
- Department of Hepatobiliary and Pancreatic Surgery, IDIBELL, Bellvitge University Hospital, University of Barcelona, L'Hospitalet de Llobregat, Spain
| | - Anna Casteràs
- Unit of Diabetes and Metabolism Research, VHIR, Vall d'Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Mar Concepción
- Department of Gastroenterology, Santa Creu i Sant Pau Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Míriam Cuatrecasas
- Department of Pathology, CIBEREHD, IDIBAPS, Hospital Clínic Barcelona, University of Barcelona, Barcelona, Spain
| | - Gloria Fernàndez Esparrach
- Unit of Endoscopy, Department of Gastroenterology, CIBEREHD IDIBAPS, Hospital Clínic Barcelona, University of Barcelona, Barcelona, Spain
| | - Esther Fort
- Department of Gastroenterology, Doctor Josep Trueta University Hospital, Girona, Spain
| | | | - Àngels Ginès
- Unit of Endoscopy, Department of Gastroenterology, CIBEREHD IDIBAPS, Hospital Clínic Barcelona, University of Barcelona, Barcelona, Spain
| | - Lucas Ilzarbe
- Department of Gastroenterology, Hospital del Mar Parc Salut Mar, Barcelona, Spain
| | - Carme Loras
- Department of Gastroenterology, CIBERehd, University of Barcelona, Terrassa, Spain
| | - Miquel Masachs
- Department of Endocopy, Vall d'Hebron University Hospital, Autonomous University of Barcelona, Terrassa, Spain
| | - Xavier Merino
- Department of Radiodiagnostic, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Jorge J Olsina
- Department of General Surgery, Institute for Research in Biomedicine of Lleida (IRBLleida), University Hospital Arnau de Vilanova, Lleida, Spain
| | - Valentí Puig-Diví
- Department of Gastroenterology, Parc Taulí Research and Innovation Institute I3PT, Parc Taulí University Hospital, Sabadell, Spain
| | - Sílvia Salord
- Unit of Hepato-Bilio-Pancreatic, Department of Digestive Diseases, IDIBELL, Bellvitge University Hospital, University of Barcelona, L'Hospitalet de Llobregat, Spain
| | - Teresa Serrano
- Department of Pathology, IDIBELL, CIBERehd, Bellvitge University Hospital, L'Hospitalet de Llobregat, Spain
| | - Eva C Vaquero
- Department of Gastroenterology, CIBEREHD IDIBAPS, Hospital Clínic Barcelona, University of Barcelona, Barcelona, Spain
| |
Collapse
|
49
|
Matsumoto T, Tanaka G, Mori S, Niki M, Sato S, Shiraki T, Iso Y, Nagashima K, Irisawa A, Nozawa Y, Takada-Owada A, Ishida K, Aoki T. A resected case of pancreatic head cancer developing 40 years after lateral pancreaticojejunostomy for chronic pancreatitis. Clin J Gastroenterol 2024; 17:537-542. [PMID: 38396137 PMCID: PMC11127812 DOI: 10.1007/s12328-024-01924-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/11/2024] [Indexed: 02/25/2024]
Abstract
A 72-year-old male patient presented to our department complaining of with upper abdominal pain and jaundice. He had a history of a side-to-side pancreaticojejunostomy performed 40 years previously for chronic pancreatitis. A diagnostic workup revealed a tumor 3 cm in size in the pancreatic head as the etiology of the jaundice. Subsequently, the patient was diagnosed with resectable pancreatic cancer. Following two cycles of neoadjuvant chemotherapy, an extended pancreatoduodenectomy was performed because of tumor invasion at the previous pancreaticojejunostomy site. Concurrent portal vein resection and reconstruction were performed. Pathological examination confirmed invasive ductal carcinoma (T2N1M0, Stage IIB). This case highlights the clinical challenges in pancreatic head carcinoma following a side-to-side pancreaticojejunostomy. Although pancreaticojejunostomy is believed to reduce the risk of pancreatic cancer in patients with chronic pancreatitis, clinicians should be aware that, even after this surgery, there is still a chance of developing pancreatic cancer during long-term follow-up.
Collapse
Affiliation(s)
- Takatsugu Matsumoto
- Department of Hepato-Biliary-Pancreatic Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan.
| | - Genki Tanaka
- Department of Hepato-Biliary-Pancreatic Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Shozo Mori
- Department of Hepato-Biliary-Pancreatic Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Maiko Niki
- Department of Hepato-Biliary-Pancreatic Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Shun Sato
- Department of Hepato-Biliary-Pancreatic Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Takayuki Shiraki
- Department of Hepato-Biliary-Pancreatic Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Yukihiro Iso
- Department of Hepato-Biliary-Pancreatic Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Kazunori Nagashima
- Department of Gastroenterology, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Atsushi Irisawa
- Department of Gastroenterology, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Yumi Nozawa
- Department of Diagnostic Pathology, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Atsuko Takada-Owada
- Department of Diagnostic Pathology, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Kazuyuki Ishida
- Department of Diagnostic Pathology, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Taku Aoki
- Department of Hepato-Biliary-Pancreatic Surgery, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| |
Collapse
|
50
|
Park JH, Mortaja M, Son HG, Zhao X, Sloat LM, Azin M, Wang J, Collier MR, Tummala KS, Mandinova A, Bardeesy N, Semenov YR, Mino-Kenudson M, Demehri S. Statin prevents cancer development in chronic inflammation by blocking interleukin 33 expression. Nat Commun 2024; 15:4099. [PMID: 38816352 PMCID: PMC11139893 DOI: 10.1038/s41467-024-48441-8] [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: 04/20/2023] [Accepted: 04/24/2024] [Indexed: 06/01/2024] Open
Abstract
Chronic inflammation is a major cause of cancer worldwide. Interleukin 33 (IL-33) is a critical initiator of cancer-prone chronic inflammation; however, its induction mechanism by environmental causes of chronic inflammation is unknown. Herein, we demonstrate that Toll-like receptor (TLR)3/4-TBK1-IRF3 pathway activation links environmental insults to IL-33 induction in the skin and pancreas inflammation. An FDA-approved drug library screen identifies pitavastatin to effectively suppress IL-33 expression by blocking TBK1 membrane recruitment/activation through the mevalonate pathway inhibition. Accordingly, pitavastatin prevents chronic pancreatitis and its cancer sequela in an IL-33-dependent manner. The IRF3-IL-33 axis is highly active in chronic pancreatitis and its associated pancreatic cancer in humans. Interestingly, pitavastatin use correlates with a significantly reduced risk of chronic pancreatitis and pancreatic cancer in patients. Our findings demonstrate that blocking the TBK1-IRF3-IL-33 signaling axis suppresses cancer-prone chronic inflammation. Statins present a safe and effective prophylactic strategy to prevent chronic inflammation and its cancer sequela.
Collapse
Affiliation(s)
- Jong Ho Park
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, South Korea
| | - Mahsa Mortaja
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Heehwa G Son
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Xutu Zhao
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lauren M Sloat
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Marjan Azin
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jun Wang
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael R Collier
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Krishna S Tummala
- Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Quantitative Biosciences, Merck Research Laboratories, Boston, MA, USA
| | - Anna Mandinova
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Nabeel Bardeesy
- Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Yevgeniy R Semenov
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Harvard Medical School, Boston, USA
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shadmehr Demehri
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|