1
|
Hu K, Zhao X, Zhang N, Ma J, Zhang R, Lu Z, Wu W, Ji Y, Li X. Effect of tumor microenvironment in pancreatic cancer on the loss of β-cell mass: implications for type 3c diabetes. J Gastroenterol 2025; 60:512-525. [PMID: 39760782 DOI: 10.1007/s00535-024-02204-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND To explore the complex interactions between the tumor microenvironment (TME) of pancreatic ductal adenocarcinoma (PDAC) and the loss of β-cell mass, further elucidating the mechanisms of type 3c diabetes mellitus (T3cDM) onset. METHODS Single-cell RNA sequencing was employed to analyze the PDAC TME, identifying cell interactions and gene expression changes of endocrine cells. Pathological changes and paraneoplastic islets were assessed in the proximal paratumor (PP) and distal paratumor (DP). Fractional β-cell area and islet density were compared among normal pancreas from donors and paraneoplastic tissues from non-diabetes mellitus (NDM) and T3cDM patients. TUNEL staining, RT-qPCR and CCK8 assay were applied to demonstrate the β-cell apoptosis. RESULTS Tumor cells, immune cells and fibroblasts could interact with endocrine cells, and apoptotic pathways were activated in endocrine cells of the PP. The PDAC TME was characterized by marked inflammation, sever fibrosis and atrophy. The islets in the PP had lower fractional β-cell area (0.68 ± 0.65% vs. 0.86 ± 1.02%, P = 0.037) and islet density (0.54 ± 0.42 counts/mm2 vs. 0.83 ± 0.90 counts/mm2, P = 0.001) compared to those in the DP. The PDAC TME in T3cDM exerted a more significant impact on the paraneoplastic islets compared to NDM. Moreover, β-cell apoptosis was markedly increased in the PP compared to the DP in PDAC patients without diabetes, particularly in smaller islets. Apoptosis-related genes were highly expressed in INS-1E cells exposed to PANC-1 medium. CONCLUSION Our research revealed that the PDAC TME is usually accompanied by some pathological changes, including inflammation, fibrosis, and atrophy. These pathological changes are related to a reduction in β-cell mass and trigger the development of T3cDM.
Collapse
Affiliation(s)
- Ke Hu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Xuelian Zhao
- Department of Pathology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Na Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Jing Ma
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Ruonan Zhang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Zhiqiang Lu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
| | - Wenchuan Wu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
| | - Xiaomu Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
2
|
Farinella R, Felici A, Peduzzi G, Testoni SGG, Costello E, Aretini P, Blazquez-Encinas R, Oz E, Pastore A, Tacelli M, Otlu B, Campa D, Gentiluomo M. From classical approaches to artificial intelligence, old and new tools for PDAC risk stratification and prediction. Semin Cancer Biol 2025; 112:71-92. [PMID: 40147701 DOI: 10.1016/j.semcancer.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is recognized as one of the most lethal malignancies, characterized by late-stage diagnosis and limited therapeutic options. Risk stratification has traditionally been performed using epidemiological studies and genetic analyses, through which key risk factors, including smoking, diabetes, chronic pancreatitis, and inherited predispositions, have been identified. However, the multifactorial nature of PDAC has often been insufficiently addressed by these methods, leading to limited precision in individualized risk assessments. Advances in artificial intelligence (AI) have been proposed as a transformative approach, allowing the integration of diverse datasets-spanning genetic, clinical, lifestyle, and imaging data into dynamic models capable of uncovering novel interactions and risk profiles. In this review, the evolution of PDAC risk stratification is explored, with classical epidemiological frameworks compared to AI-driven methodologies. Genetic insights, including genome-wide association studies and polygenic risk scores, are discussed, alongside AI models such as machine learning, radiomics, and deep learning. Strengths and limitations of these approaches are evaluated, with challenges in clinical translation, such as data scarcity, model interpretability, and external validation, addressed. Finally, future directions are proposed for combining classical and AI-driven methodologies to develop scalable, personalized predictive tools for PDAC, with the goal of improving early detection and patient outcomes.
Collapse
Affiliation(s)
| | | | | | - Sabrina Gloria Giulia Testoni
- Division of Gastroenterology and Gastrointestinal Endoscopy, IRCCS Policlinico San Donato, Vita-Salute San Raffaele University, Milan, Italy
| | - Eithne Costello
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Paolo Aretini
- Fondazione Pisana per la Scienza, San Giuliano Terme, Italy
| | - Ricardo Blazquez-Encinas
- Department of Cell Biology, Physiology and Immunology, University of Cordoba / Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Elif Oz
- Department of Biostatistics and Bioinformatics, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Aldo Pastore
- Fondazione Pisana per la Scienza, San Giuliano Terme, Italy
| | - Matteo Tacelli
- Pancreas Translational & Clinical Research Center, Pancreato-Biliary Endoscopy and Endosonography Division, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Burçak Otlu
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, Ankara, Turkey
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | | |
Collapse
|
3
|
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 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] [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
|
4
|
Mohindroo C, Dy PS, Hande SP, D'Adamo CR, Mavanur A, Thomas A, McAllister F, De Jesus-Acosta A. New onset diabetes predicts clinical outcomes in patients with pancreatic adenocarcinoma. J Gastrointest Oncol 2025; 16:226-233. [PMID: 40115918 PMCID: PMC11921277 DOI: 10.21037/jgo-24-570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/12/2024] [Indexed: 03/23/2025] Open
Abstract
Background One percent of pancreatic adenocarcinoma (PDAC) patients are diagnosed with new onset diabetes (NOD) over the age of 50 years within 3 years. Therefore, NOD is a major factor for early diagnosis of PDAC. Research has focused on understanding the differences between NOD and type 2 diabetes, particularly in relation to PDAC. However, conflicting data exists regarding their impact on survival outcomes in PDAC patients. We performed this multi-center study to assess the prevalence and influence of NOD on clinical outcomes in patients with PDAC within a community-based hospital system. Methods We conducted a retrospective cohort study of 138 patients with biopsy-proven PDAC with localized/borderline disease (n=70), and metastatic disease (n=68) at three institutions from 2014 to 2021. NOD group consisted of pts diagnosed with diabetes [hemoglobin A1c (HbA1c) >6.5%] or pre-diabetes (HbA1c 5.7-6.4%) within the 3 years prior to PDAC diagnosis. Primary aim of the study was to determine the impact of NOD on clinical outcomes. Results A total of 138 patients were included in the study, from which 30 met the criteria for NOD. No significant differences were noted in the demographic and clinical characteristics comparing patients based on NOD history. Comparing survival outcomes, NOD group was associated with worse overall survival (OS) in both the metastatic cohort [n=68, progression-free survival (PFS) 4.6 vs. 7.1 months, P=0.07; OS 7.1 vs. 13.2 months, P=0.01) and the resected cohort (n=40, PFS 8.4 vs. 19.3 months, P=0.04; OS 24.5 vs. 42.3 months, P=0.04). In multivariate analysis, the impact of NOD remained significant for OS and PFS in the resected cohort. Identifying common features amongst the NOD group, we found the entire cohort had a significant reduction in individual body mass index (BMI) 1 year prior to the NOD diagnosis (P=0.006). Conclusions NOD is associated with worse survival outcomes in patients with metastatic and resected PDAC. Reduction of BMI prior to diagnosis of NOD, warrants further investigation to be incorporated into the PDAC screening paradigm.
Collapse
Affiliation(s)
- Chirayu Mohindroo
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Paul S Dy
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Suraj P Hande
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Christopher R D'Adamo
- Department of Family and Community Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Arun Mavanur
- Department of Surgery, Sinai Hospital of Baltimore, Baltimore, MD, USA
- Department of Surgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Asha Thomas
- Division of Endocrinology, Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Florencia McAllister
- Departments of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ana De Jesus-Acosta
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
5
|
Hu C, Chen Y, Yin X, Xu R, Yin C, Wang C, Zhao Y. Pancreatic endocrine and exocrine signaling and crosstalk in physiological and pathological status. Signal Transduct Target Ther 2025; 10:39. [PMID: 39948335 PMCID: PMC11825823 DOI: 10.1038/s41392-024-02098-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/20/2024] [Accepted: 12/03/2024] [Indexed: 02/16/2025] Open
Abstract
The pancreas, an organ with dual functions, regulates blood glucose levels through the endocrine system by secreting hormones such as insulin and glucagon. It also aids digestion through the exocrine system by secreting digestive enzymes. Complex interactions and signaling mechanisms between the endocrine and exocrine functions of the pancreas play a crucial role in maintaining metabolic homeostasis and overall health. Compelling evidence indicates direct and indirect crosstalk between the endocrine and exocrine parts, influencing the development of diseases affecting both. From a developmental perspective, the exocrine and endocrine parts share the same origin-the "tip-trunk" domain. In certain circumstances, pancreatic exocrine cells may transdifferentiate into endocrine-like cells, such as insulin-secreting cells. Additionally, several pancreatic diseases, including pancreatic cancer, pancreatitis, and diabetes, exhibit potential relevance to both endocrine and exocrine functions. Endocrine cells may communicate with exocrine cells directly through cytokines or indirectly by regulating the immune microenvironment. This crosstalk affects the onset and progression of these diseases. This review summarizes the history and milestones of findings related to the exocrine and endocrine pancreas, their embryonic development, phenotypic transformations, signaling roles in health and disease, the endocrine-exocrine crosstalk from the perspective of diseases, and potential therapeutic targets. Elucidating the regulatory mechanisms of pancreatic endocrine and exocrine signaling and provide novel insights for the understanding and treatment of diseases.
Collapse
Grants
- National High Level Hospital Clinical Research Funding (2022, 2022-PUMCH-D-001, to YZ), CAMS Innovation Fund for Medical Sciences (2021, 2021-I2M-1-002, to YZ), National Nature Science Foundation of China (2021, 82102810, to CW, the Fundamental Research Funds for the Central Universities(3332023123)
- cNational High Level Hospital Clinical Research Funding (2022, 2022-PUMCH-D-001, to YZ), CAMS Innovation Fund for Medical Sciences (2021, 2021-I2M-1-002, to YZ), National Nature Science Foundation of China (2021, 82102810, to CW, the Fundamental Research Funds for the Central Universities(3332023123)
Collapse
Affiliation(s)
- Chenglin Hu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Yuan Chen
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Xinpeng Yin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Chenxue Yin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China
| | - Chengcheng Wang
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China.
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, PR China.
- Institute of Clinical Medicine, Peking Union Medical College Hospital, Beijing, PR China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- State Key Laboratory of Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, PR China.
- National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Beijing, PR China.
| |
Collapse
|
6
|
Heller M, Mann DA, Katona BW. Current Approaches of Pancreatic Cancer Surveillance in High-Risk Individuals. J Gastrointest Cancer 2025; 56:61. [PMID: 39932614 DOI: 10.1007/s12029-025-01184-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2025] [Indexed: 02/14/2025]
Abstract
Currently, those recommended to undergo pancreatic cancer (PC) surveillance include appropriately aged individuals at high risk of PC due to an identifiable genetic susceptibility or those without identifiable genetic susceptibility who nonetheless have a strong family history of PC. With increases in identification of individuals at high risk for PC and increased use of PC surveillance in clinical practice, there has been increasing debate about who should undergo surveillance as well as how surveillance should be performed including use of imaging and blood-based testing. Furthermore, there is increasing interest in the outcomes of PC surveillance in high-risk individuals with some studies demonstrating that surveillance leads to downstaging of PC and improvements in survival. In this review, we summarize the current state of PC surveillance in high-risk individuals, providing an overview of the risk factors associated with PC, selection of high-risk individuals for PC surveillance, and the current, but non-uniform, recommendations for performing PC surveillance. Additionally, we review approaches to apply various imaging and blood-based tests to surveillance and the outcomes of PC surveillance.
Collapse
Affiliation(s)
- Melissa Heller
- Division of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Derek A Mann
- Division of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Bryson W Katona
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd., 751 South Pavilion, Philadelphia, PA, 19104, USA.
| |
Collapse
|
7
|
Solaiyappan M, Bharti SK, Sharma RK, Dbouk M, Nizam W, Brock MV, Goggins MG, Bhujwalla ZM. Artificial neural network detection of pancreatic cancer from proton (1H) magnetic resonance spectroscopy patterns of plasma metabolites. COMMUNICATIONS MEDICINE 2025; 5:24. [PMID: 39838068 PMCID: PMC11751387 DOI: 10.1038/s43856-024-00727-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Routine screening to detect silent but deadly cancers such as pancreatic ductal adenocarcinoma (PDAC) can significantly improve survival, creating an important need for a convenient screening test. High-resolution proton (1H) magnetic resonance spectroscopy (MRS) of plasma identifies circulating metabolites that can allow detection of cancers such as PDAC that have highly dysregulated metabolism. METHODS We first acquired 1H MR spectra of human plasma samples classified as normal, benign pancreatic disease and malignant (PDAC). We next trained a system of artificial neural networks (ANNs) to process and discriminate these three classes using the full spectrum range and resolution of the acquired spectral data. We then identified and ranked spectral regions that played a salient role in the discrimination to provide interpretability of the results. We tested the accuracy of the ANN performance using blinded plasma samples. RESULTS We show that our ANN approach yields, in a cross validation-based training of 170 samples, a sensitivity and a specificity of 100% for malignant versus non-malignant (normal and disease combined) discrimination. The trained ANNs achieve a sensitivity and specificity of 87.5% and 93.1% respectively (AUC: ROC = 0.931, P-R = 0.854), with 45 blinded plasma samples. Further, we show that the salient spectral regions of the ANN discrimination correspond to metabolites of known importance for their role in cancers. CONCLUSIONS Our results demonstrate that the ANN approach presented here can identify PDAC from 1H MR plasma spectra to provide a convenient plasma-based assay for population-level screening of PDAC. The ANN approach can be suitably expanded to detect other cancers with metabolic dysregulation.
Collapse
Affiliation(s)
- Meiyappan Solaiyappan
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Santosh Kumar Bharti
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Raj Kumar Sharma
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mohamad Dbouk
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wasay Nizam
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Malcolm V Brock
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael G Goggins
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zaver M Bhujwalla
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
8
|
Cencioni C, Malatesta S, Vigiano Benedetti V, Licursi V, Perfetto L, Conte F, Ranieri D, Bartolazzi A, Kunkl M, Tuosto L, Larghi A, Piro G, Agostini A, Tortora G, Corbo V, Carbone C, Spallotta F. The GLP-1R agonist semaglutide reshapes pancreatic cancer associated fibroblasts reducing collagen proline hydroxylation and favoring T lymphocyte infiltration. J Exp Clin Cancer Res 2025; 44:18. [PMID: 39828692 PMCID: PMC11744909 DOI: 10.1186/s13046-024-03263-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Metabolic syndrome represents a pancreatic ductal adenocarcinoma (PDAC) risk factor. Metabolic alterations favor PDAC onset, which occurs early upon dysmetabolism. Pancreatic neoplastic lesions evolve within a dense desmoplastic stroma, consisting in abundant extracellular matrix settled by cancer associated fibroblasts (CAFs). Hereby, dysmetabolism and PDAC association was analyzed focusing on CAF functions. METHODS PDAC development upon dysmetabolic conditions was investigated in: 1) high fat diet fed wild type immunocompetent syngeneic mice by orthotopic transplantation of pancreatic intraepithelial neoplasia (PanIN) organoids; and 2) primary pancreatic CAFs isolated from chemotherapy naïve PDAC patients with/without an history of metabolic syndrome. RESULTS The dysmetabolic-associated higher PDAC aggressiveness was paralleled by collagen fibril enrichment due to prolyl 4-hydroxylase subunit alpha 1 (P4HA1) increased function. Upon dysmetabolism, P4HA1 boosts collagen proline hydroxylation, intensifies collagen contraction strength, precluding PDAC infiltration. Noteworthy, semaglutide, an incretin agonist, prevents the higher dysmetabolism-dependent PDAC stromal deposition and allows T lymphocyte infiltration, reducing tumor development. CONCLUSIONS These results shed light on novel therapeutic options for PDAC patients with metabolic syndrome aimed at PDAC stroma reshape.
Collapse
Affiliation(s)
- Chiara Cencioni
- Institute of System Analysis and Informatics "Antonio Ruberti", National Research Council (IASI-CNR), 00185, Rome, Italy
| | - Silvia Malatesta
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185, Rome, Italy
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185, Rome, Italy
| | | | - Valerio Licursi
- Institute of Molecular Biology and Pathology, National Research Council (IBPM-CNR), 00185, Rome, Italy
| | - Livia Perfetto
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185, Rome, Italy
| | - Federica Conte
- Institute of System Analysis and Informatics "Antonio Ruberti", National Research Council (IASI-CNR), 00185, Rome, Italy
| | - Danilo Ranieri
- Dipartimento Di Scienze Della Vita, Della Salute E Delle Professioni Sanitarie. Università Degli Studi "Link Campus University", 00165, Rome, Italy
| | - Armando Bartolazzi
- Pathology Research Laboratory, Sant' Andrea University Hospital, 00189, Rome, Italy
| | - Martina Kunkl
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185, Rome, Italy
- Neuroimmunology Unit, IRCCS Santa Lucia Foundation, 00179, Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185, Rome, Italy
| | - Alberto Larghi
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
- Center for Endoscopic Research Therapeutics and Training (CERTT), Catholic University, 00168, Rome, Italy
| | - Geny Piro
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | - Antonio Agostini
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | - Giampaolo Tortora
- Department of Translational Medicine, Catholic University of the Sacred Heart, 00168, Rome, Italy
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine (DIMI), University and Hospital Trust of Verona, 37100, Verona, Italy
| | - Carmine Carbone
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | - Francesco Spallotta
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University, 00185, Rome, Italy.
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185, Rome, Italy.
| |
Collapse
|
9
|
Zhao X, Wang Y, Zhou L, Ye A, Zhu Q. Changes of CA19-9 levels and related influencing factors in patients with type 2 diabetes mellitus after antidiabetic therapy. Sci Rep 2025; 15:1264. [PMID: 39779798 PMCID: PMC11711652 DOI: 10.1038/s41598-025-85807-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025] Open
Abstract
Abnormalities of carbohydrate antigen 19 - 9 (CA19-9) are common in patients with type 2 diabetes mellitus (T2DM), and in some patients, CA19-9 returns to normal level after glycemic control. The aim of this study was to investigate the factors associated with CA19-9 levels in patients with T2DM and the associated influences on the degree of reduction of CA19-9 levels after antidiabetic therapy (AT). This study was an observational cross-sectional study. A total of 213 patients with T2DM were enrolled in this study, of whom 105 with abnormal CA19-9 and 108 with normal CA19-9. Socio-demographic information, complete blood counts, biochemical indicators, thyroid function indicators, and CA19-9 level were collected separately for each subject. Levels of glycosylated hemoglobin, type A1C (HbA1c), fasting blood glucose (FBG) were significantly higher in T2DM patients with abnormal CA19-9 compared to patients with normal CA19-9 (both FDR < 0.001). CA19-9 level was significantly and positively correlated with neutrophil/lymphocyte ratio (NLR) (r = 0.16, P = 0.02), monocyte/lymphocyte ratio (MLR) (r = 0.16, P = 0.02), and FBG (r = 0.38, P < 0.001), while significantly and negatively correlated with free triiodothyronine (FT3) (r=-0.22, P = 0.002) and albumin count (r=-0.18, P = 0.007). After AT, the degree of decrease in CA19-9 level in T2DM patients with abnormal CA19-9 was significantly positively correlated with degree of decrease in FBG (r = 0.33, P < 0.001), as well as CA19-9 level before AT (r = 0.73, P < 0.001), NLR (r = 0.20, P = 0.04), and MLR (r = 0.25, P = 0.01). In this study, we investigated the influencing factors associated with CA19-9 level and the factors influencing degree of CA19-9 reduction after AT in T2DM patients with abnormal CA19-9.
Collapse
Affiliation(s)
- Xiafei Zhao
- Affiliated Xiaoshan Hospital, Hangzhou Normal University/Zhejiang Xiaoshan Hospital, 728 Yucai North Road, Hangzhou, 311200, China
| | - Yan Wang
- Affiliated Xiaoshan Hospital, Hangzhou Normal University/Zhejiang Xiaoshan Hospital, 728 Yucai North Road, Hangzhou, 311200, China
| | - Ling Zhou
- Affiliated Xiaoshan Hospital, Hangzhou Normal University/Zhejiang Xiaoshan Hospital, 728 Yucai North Road, Hangzhou, 311200, China
| | - Aili Ye
- Affiliated Xiaoshan Hospital, Hangzhou Normal University/Zhejiang Xiaoshan Hospital, 728 Yucai North Road, Hangzhou, 311200, China.
| | - Quanfeng Zhu
- Affiliated Xiaoshan Hospital, Hangzhou Normal University/Zhejiang Xiaoshan Hospital, 728 Yucai North Road, Hangzhou, 311200, China.
| |
Collapse
|
10
|
Elfekih H, Charfeddine Y, Said MA, Saafi W, Jaziri H, Mraidha MH, Halloul I, Ben Ali A, Yacoub S, Brahem S, Guesmi A, Ksiaa M, Ben Ahmed S, Sahli J, Saad G, Hasni Y. Comparative Analysis of Long-Standing and Newly Diagnosed Diabetes Mellitus in Patients with Pancreatic Ductal Adenocarcinoma: A Tunisian Multicenter Study. LA TUNISIE MEDICALE 2025; 103:112-116. [PMID: 39812203 PMCID: PMC11906228 DOI: 10.62438/tunismed.v103i1.5328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/23/2024] [Indexed: 01/16/2025]
Abstract
INTRODUCTION Diabetes mellitus has emerged as a global public health issue due to its increasing prevalence and the increased risk of developing cancers. Pancreatic cancer is believed to be both a consequence of pre-existing diabetes and a potential cause of new-onset diabetes. AIM This study aims to compare the characteristics of patients with pancreatic ductal adenocarcinoma and newly diagnosed or long-standing diabetes mellitus. METHODS A multicentric retrospective study was conducted over 13 years at two university hospitals in Sousse, Tunisia. Included patients had whether a newly diagnosed or a long-standing diabetes mellitus with histologically confirmed pancreatic ductal adenocarcinoma. Statistical analysis using appropriate tests was conducted. RESULTS The prevalence of diabetes mellitus was 44.6% among three hundred and seven patients with pancreatic cancer. The male-to-female ratio in patients with pancreatic ductal adenocarcinoma was 2.6:1. Patients' mean age was 63.9 years, with the majority being over 50 years old. Most patients had no family history of diabetes and exhibited significant weight loss, low body mass index, and uncontrolled diabetes. The comparison between individuals with newly diagnosed diabetes and those with long-standing diabetes revealed numerous similarities, apart from significant differences in drinking patterns (p = 0.03), tumor size (p = 0.018), and smoking in the subgroup of males (p = 0.044). CONCLUSION Patients over 50 with newly diagnosed diabetes mellitus, particularly those who consume alcohol occasionally and men who are not heavy smokers, should undergo further evaluation to identify potential early-stage pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Hamza Elfekih
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Endocrinology Diabetology, 4000, Sousse, Tunisia
| | - Yasmine Charfeddine
- University of Sousse, Faculty of Medicine of Sousse, Department of Family and Community Medicine, 4002, Sousse, Tunisia
| | - Mohamed Amine Said
- University of Sousse, Faculty of Medicine of Sousse, 4002, Sahloul University Hospital, Department of Surgery, 4054, Sousse, Tunisia
| | - Wiem Saafi
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Endocrinology Diabetology, 4000, Sousse, Tunisia
| | - Hanen Jaziri
- University of Sousse, Faculty of Medicine of Sousse, 4002, Sahloul University Hospital, Department of Gastroenterology, 4054, Sousse, Tunisia
| | - Mohamed Hedi Mraidha
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Surgery, 4000, Sousse, Tunisia
| | - Imen Halloul
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Endocrinology Diabetology, 4000, Sousse, Tunisia
| | - Azer Ben Ali
- University of Sousse, Faculty of Medicine of Sousse, 4002, Sahloul University Hospital, Department of Surgery, 4054, Sousse, Tunisia
| | - Sarra Yacoub
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Pathology, 4000, Sousse, Tunisia
| | - Salem Brahem
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Radiology, 4000, Sousse, Tunisia
| | - Ayoub Guesmi
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Radiology, 4000, Sousse, Tunisia
| | - Mehdi Ksiaa
- University of Sousse, Faculty of Medicine of Sousse, 4002, Sahloul University Hospital, Department of Gastroenterology, 4054, Sousse, Tunisia
| | - Slim Ben Ahmed
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Medical Oncology, 4000, Sousse, Tunisia
| | - Jihen Sahli
- University of Sousse, Faculty of Medicine of Sousse, Department of Family and Community Medicine, 4002, Sousse, Tunisia
| | - Ghada Saad
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Endocrinology Diabetology, 4000, Sousse, Tunisiaa
| | - Yosra Hasni
- University of Sousse, Faculty of Medicine of Sousse, 4002, Farhat Hached University Hospital, Department of Endocrinology Diabetology, 4000, Sousse, Tunisia
| |
Collapse
|
11
|
Trembath HE, LaBella ME, Kearney JF, Hariharan A, Zarmer S, Nabors M, McCabe I, Zhao RT, Meyers M, Kim HJ, Yeh JJ. New Onset Diabetes in Pancreatic Adenocarcinoma Does Not Correlate With Molecular Subtype. J Surg Oncol 2024. [PMID: 39711008 DOI: 10.1002/jso.28044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 11/23/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND AND OBJECTIVES Studies show that new onset diabetes mellitus (DM) (NOD) predates the diagnosis of PDAC by up to 2 years. Two tumor-intrinsic molecular subtypes of PDAC that are prognostic and predictive of chemotherapy response have been described and validated. We hypothesize that patients with NOD may have different molecular subtypes and prognoses. METHODS This is a single-institution study of patients who underwent resection for PDAC from 2009 to 2022 with de-identified samples available for sequencing. Demographic and clinical factors were examined using bivariate and multivariate analysis. RESULTS A total of 97 patients met inclusion criteria: 70 with no history of DM, 11 with longstanding DM (> 2 years), and 16 with NOD. The demographics between groups were overall similar. After controlling for age, sex, race, BMI, and tobacco history, NOD was not a significant predictor of PDAC subtype. There were no survival differences between groups. Transcriptomic analysis suggests the upregulation of inflammatory and immune activation and regulation pathways in NOD. CONCLUSIONS As continued interest in NOD and PDAC mounts, we are the first to examine if NOD may be associated with molecular subtypes and outcomes. Further investigation into the underlying pathophysiology of the NOD group is still needed.
Collapse
Affiliation(s)
- Hannah E Trembath
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michelle E LaBella
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joseph F Kearney
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Arthi Hariharan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sandra Zarmer
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mariaelena Nabors
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ian McCabe
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ryan T Zhao
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael Meyers
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Hong Jin Kim
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jen Jen Yeh
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
12
|
Meziani J, de Jong JGY, Fuhler GM, Koopmann BDM, Levink IJM, Fockens P, Vleggaar FP, Bruno MJ, Cahen DL. Assessment of Glucose and HbA1c Monitoring in a Pancreatic Cancer Surveillance Program for High-Risk Individuals. Clin Transl Gastroenterol 2024; 15:e00777. [PMID: 39413349 DOI: 10.14309/ctg.0000000000000777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
INTRODUCTION Several studies suggest that new-onset diabetes mellitus is an early manifestation of pancreatic ductal adenocarcinoma (PDAC). Therefore, the International Cancer of the Pancreas Screening Consortium recommends glucose and hemoglobin A1c (HbA1c) monitoring in high-risk individuals (HRIs) undergoing surveillance. However, evidence that such monitoring improves PDAC detection is lacking. Our aim was to investigate the association between serum glucose and HbA1c values and the development of PDAC in HRIs undergoing surveillance. METHODS Participants were recruited from the familial pancreatic cancer surveillance cohort, which follows hereditary predisposed HRIs yearly by magnetic resonance imaging and/or endoscopic ultrasound and blood sampling. Those who underwent fasting glucose and/or HbA1c monitoring at least once were eligible candidates. RESULTS Four hundred four HRIs met the inclusion criteria. During a median follow-up of 41 months (range 14-120), 9 individuals developed PDAC and 4 (without PDAC) were diagnosed with new-onset diabetes mellitus. Glucose levels ranged from 3.4 to 10.7 mmol/L (mean 5.6 ± 0.7) and HbA1c levels from 25 to 68 mmol/mol (mean 37.7 ± 4.1). The mean values did not differ significantly between PDAC cases and controls. The percentage of individuals with at least one elevated value were comparable between PDAC cases and controls for glucose (33% and 27%, P = 0.707) and HbA1c (22% and 14%, P = 0.623). No consistent glucose or HbA1c trends over time suggested a correlation with PDAC development. DISCUSSION In this HRI surveillance cohort, measuring glucose and HbA1c values did not contribute to PDAC detection. Larger and longer-term studies are needed to determine the final role of glucose and HbA1c monitoring in PDAC surveillance.
Collapse
Affiliation(s)
- Jihane Meziani
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jedidja G Y de Jong
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Gwenny M Fuhler
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Brechtje D M Koopmann
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Iris J M Levink
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Paul Fockens
- Department of Gastroenterology & Hepatology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Frank P Vleggaar
- Department of Gastroenterology & Hepatology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marco J Bruno
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Djuna L Cahen
- Department of Gastroenterology & Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
13
|
Zhu X, Xiao Z, Liu H, Zhang P, Deng S, Ding L, Feng J, Luo J, Ni Q, Luo G, Yu X. Pancreatic Cancer: An Exocrine Tumor With Endocrine Characteristics. Ann Surg 2024; 280:e17-e25. [PMID: 38050737 PMCID: PMC11542970 DOI: 10.1097/sla.0000000000006168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
OBJECTIVE To examine the characteristics of patients with pancreatic cancer with long-term survival. BACKGROUND Although pancreatic cancer is a highly lethal malignancy, a minority of patients experience long-term survival. The characteristics of these patients remain largely unidentified. METHODS An indolent subgroup was established using carbohydrate antigen 19-9 (CA19-9), which is the best-validated biomarker for pancreatic cancer. Of 1558 patients, 13.9% were included in the CA19-9-normal (≤37 U/mL) subgroup. RESULTS A normal CA19-9 level was an independent variable for overall survival (median survival, 18.1 vs 9.7 months, hazard ratio = 0.53, P < 0.001). The 5-year survival of patients with stage IV CA19-9-normal cancer was higher than that of patients with stage I-IV CA19-9-high cancer (22.4% vs 6.8%, P = 0.034). The CA19-9-normal subgroup exhibited reduced levels of circulating glucose ( P < 0.001) and increased expression of insulin ( P < 0.001) compared with the CA19-9-high subgroup. Glucose was a substrate for CA19-9 biosynthesis through the hexosamine biosynthesis pathway. In addition, in pancreatic cancer animal models of diabetes, glucose control decreased CA19-9 levels and improved overall survival. In a clinical trial (NCT05306028) of patients before undergoing major anticancer treatments, glucose control decreased CA19-9 levels in 90.9% of the patients. CONCLUSIONS CA19-9-normal pancreatic cancer is a strikingly indolent subgroup with low glucose and high insulin. Glucose control is a promising therapeutic strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Xinzhe Zhu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China
- Department of Oncology, Shanghai Medical College, Fudan University, China
- Shanghai Pancreatic Cancer Institute, China
- Pancreatic Cancer Institute, Fudan University, China
| | - Zhiwen Xiao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China
- Department of Oncology, Shanghai Medical College, Fudan University, China
- Shanghai Pancreatic Cancer Institute, China
- Pancreatic Cancer Institute, Fudan University, China
| | - He Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China
- Department of Oncology, Shanghai Medical College, Fudan University, China
- Shanghai Pancreatic Cancer Institute, China
- Pancreatic Cancer Institute, Fudan University, China
| | - Pin Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China
- Department of Oncology, Shanghai Medical College, Fudan University, China
- Shanghai Pancreatic Cancer Institute, China
- Pancreatic Cancer Institute, Fudan University, China
| | - Shengming Deng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China
- Department of Oncology, Shanghai Medical College, Fudan University, China
- Shanghai Pancreatic Cancer Institute, China
- Pancreatic Cancer Institute, Fudan University, China
| | - Lei Ding
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China
- Department of Oncology, Shanghai Medical College, Fudan University, China
- Shanghai Pancreatic Cancer Institute, China
- Pancreatic Cancer Institute, Fudan University, China
| | - Jingjing Feng
- Department of Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jianfeng Luo
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
| | - Quanxing Ni
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China
- Department of Oncology, Shanghai Medical College, Fudan University, China
- Shanghai Pancreatic Cancer Institute, China
- Pancreatic Cancer Institute, Fudan University, China
| | - Guopei Luo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China
- Department of Oncology, Shanghai Medical College, Fudan University, China
- Shanghai Pancreatic Cancer Institute, China
- Pancreatic Cancer Institute, Fudan University, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, China
- Department of Oncology, Shanghai Medical College, Fudan University, China
- Shanghai Pancreatic Cancer Institute, China
- Pancreatic Cancer Institute, Fudan University, China
| |
Collapse
|
14
|
Cho SH, Lee Y, Huh G, Jeong H, Yoo C, Tae Jun S, Seo DW, Oh D. Acute pancreatitis as an early sign of pancreatic cancer; a retrospective, matched cohort study. Scand J Gastroenterol 2024; 59:1330-1335. [PMID: 39387458 DOI: 10.1080/00365521.2024.2414804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/30/2024] [Accepted: 10/06/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND AND AIMS Pancreatic ductal adenocarcinoma (PDAC) often presents as acute pancreatitis (AP). However, data on the clinical outcomes of PDAC initially presenting as AP are limited. We aimed to assess the clinical features of PDAC that manifest as AP. METHODS We reviewed the PDAC database at the Asan Medical Center between 2010-2016. Our study included 77 patients with PDAC who presented with AP (PDAC-AP group) and 154 age-gender-matched PDAC patients as controls (PDAC-other group). Patients' demographics, disease characteristics, and outcomes were compared between both groups. RESULTS Acute pancreatitis was an initial symptom in 1.12% of the patients with PDAC (77 of 6,821). Approximately 81.8% of the patients had clinically mild pancreatitis, and 91% were diagnosed with PDAC within two months of presentation with AP. Main tumor size was significantly smaller in the PDAC-AP group than in the PDAC-other group (PDAC-AP: 2.59 ± 1.21 cm vs. PDAC-other: 3.73 ± 1.78 cm, p < 0.01). The PDAC-AP group patients were diagnosed earlier than those in the PDAC-other group (PDAC-AP: stage 1-2, 80.6% vs. PDAC-other: 46.7%, p < 0.01). The proportion of resectable PDAC was significantly higher in the PDAC-AP group (PDAC-AP: 64.9% vs. PDAC-other: 50%, p < 0.01). Overall survival was significantly longer in the PDAC-AP group than in the PDAC-other group (30.2 months vs. 19.9 months, p = 0.03). CONCLUSIONS In patients who presented with clinical AP, PDAC was identified at an earlier stage, and these patients showed better survival rates. These results suggest that AP may be an early sign of PDAC.
Collapse
Affiliation(s)
- Sung Hyun Cho
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yoonchan Lee
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Gunn Huh
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hyehyun Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Song Tae Jun
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Dong-Wan Seo
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Dongwook Oh
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
15
|
Lee GH, Kim YH, Woo SM, Lee WJ, Han SS, Park SJ, Price S, Tembo P, Hébert JR, Kim MK. The Impact of the Dietary Inflammatory Index, Fasting Blood Glucose, and Smoking Status on the Incidence and Survival of Pancreatic Cancer: A Retrospective Case-Control Study and a Prospective Study. Nutrients 2024; 16:3941. [PMID: 39599726 PMCID: PMC11597200 DOI: 10.3390/nu16223941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Pancreatic cancer (PC), a highly malignant cancer with a poor diagnosis, may be influenced by diet-related inflammation. This study examined the association between dietary inflammatory index (DII) scores and the incidence and prognosis of PC in Korea. METHODS A total of 55 patients with PC were matched with 280 healthy controls (HCs) by age and sex. We also analyzed the combined effects of DII scores and fasting blood glucose (FBG) levels or smoking status on the risk of PC and performed a survival analysis using the Cox proportional hazards method. RESULTS The DII scores were higher in the patients with PC than those in HCs (odds ratio [OR] = 3.36, confidence interval [CI] = 1.16-9.73, p = 0.03), and the effect was larger in women (OR = 6.13, CI = 1.11-33.82, p = 0.04). A high DII score was jointly associated with FBG ≥ 126 mg/dL in raising PC risk [OR = 32.5, relative excess risk due to interaction/synergy (RERI/S) index = 24.2/4.34, p-interaction = 0.04], indicating a multiplicative interaction. A high DII score combined with ex/current smoker status increased PC risk through an additive interaction (RERI/S = 1.01/1.54, p-interaction = 0.76). However, DII scores did not influence disease-free survival. CONCLUSIONS The consumption of an anti-inflammatory diet, coupled with maintaining normal FBG levels and abstaining from smoking, may help reduce the risk of PC by mitigating pancreatic inflammation.
Collapse
Affiliation(s)
- Ga Hyun Lee
- Cancer Epidemiology Branch, Division of Cancer Epidemiology and Prevention, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Republic of Korea; (G.H.L.); (Y.H.K.)
| | - Yeon Hee Kim
- Cancer Epidemiology Branch, Division of Cancer Epidemiology and Prevention, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Republic of Korea; (G.H.L.); (Y.H.K.)
| | - Sang Myung Woo
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Republic of Korea; (S.M.W.); (W.J.L.); (S.-S.H.); (S.-J.P.)
| | - Woo Jin Lee
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Republic of Korea; (S.M.W.); (W.J.L.); (S.-S.H.); (S.-J.P.)
| | - Sung-Sik Han
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Republic of Korea; (S.M.W.); (W.J.L.); (S.-S.H.); (S.-J.P.)
| | - Sang-Jae Park
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Republic of Korea; (S.M.W.); (W.J.L.); (S.-S.H.); (S.-J.P.)
| | - Sherry Price
- Department of Epidemiology and Biostatistics and Cancer Prevention and Control Program, University of South Carolina, Columbia, SC 29208, USA; (S.P.); (P.T.); (J.R.H.)
| | - Penias Tembo
- Department of Epidemiology and Biostatistics and Cancer Prevention and Control Program, University of South Carolina, Columbia, SC 29208, USA; (S.P.); (P.T.); (J.R.H.)
| | - James R. Hébert
- Department of Epidemiology and Biostatistics and Cancer Prevention and Control Program, University of South Carolina, Columbia, SC 29208, USA; (S.P.); (P.T.); (J.R.H.)
- Department of Nutrition, Connecting Health Innovations LLC, Columbia, SC 29201, USA
| | - Mi Kyung Kim
- Cancer Epidemiology Branch, Division of Cancer Epidemiology and Prevention, National Cancer Center, Ilsandong-gu, Goyang-si 10408, Republic of Korea; (G.H.L.); (Y.H.K.)
| |
Collapse
|
16
|
Wlodarczyk B, Durko L, Walczak K, Talar-Wojnarowska R, Malecka-Wojciesko E. Select Endocrine Disorders and Exosomes in Early PDAC Diagnosis. Int J Mol Sci 2024; 25:12159. [PMID: 39596226 PMCID: PMC11594802 DOI: 10.3390/ijms252212159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/01/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
Disturbances in carbohydrate metabolism are suggested to be the early symptoms of pancreatic ductal adenocarcinoma (PDAC). The accumulated data suggests that endocrine function-related biomarkers may represent a breakthrough in the early detection of PDAC. Factors which may predispose one to the development of PDAC are insulin resistance and hyperinsulinemia. Elevated insulin levels induce the onset of carcinogenesis by altering the differentiation and function of islet cells through stimulating growth factors, including insulin-like growth factors (IGFs). Impaired β cell function, along with the impact of PDAC-released factors (e.g., adrenomedullin (ADM), IGF-1, and macrophage inhibitory factor (MIF) on pancreatic islets, may contribute to the induction of diabetes associated with PDAC. Recently, exosomes have attracted worldwide attention due to their role in varied features of cell function, particularly in cancer progression. Exosomes comprise of small extracellular vesicles produced by almost all cells. These vesicles contain a vast array of biomolecules, including proteins and microRNAs. Exosomes participate in cancer growth and promote angiogenesis. They promote tumorigenesis and metastasis, and are associated with the acquisition of cancer cells resistant to chemotherapy. Data have been accumulating recently on the role of exosomes in the rapid recognition, prognosis and potential therapy of pancreatic cancer.
Collapse
Affiliation(s)
- Barbara Wlodarczyk
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-153 Lodz, Poland
| | - Lukasz Durko
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-153 Lodz, Poland
| | - Konrad Walczak
- Department of Internal Diseases and Nephrodiabetology, Medical University of Lodz, 90-549 Lodz, Poland
| | | | - Ewa Malecka-Wojciesko
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-153 Lodz, Poland
| |
Collapse
|
17
|
Le MV, Fourlanos S, Barmanray RD. Diabetic ketoacidosis as first presentation of undiagnosed pancreatic cancer in an octogenarian. Endocrinol Diabetes Metab Case Rep 2024; 2024:23-0145. [PMID: 39475781 PMCID: PMC11558915 DOI: 10.1530/edm-23-0145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 10/02/2024] [Indexed: 11/15/2024] Open
Abstract
Summary Diabetic ketoacidosis (DKA) is a complication of diabetes mellitus (DM) that can theoretically occur in people of any age. While DKA can typically be the first presentation of type 1 DM in younger people, a first presentation is rare in older adults. Pancreatic cancer often manifests with new DM or hyperglycaemia, but very rarely as DKA. We report a case of an 89-year-old woman who was incidentally diagnosed with DKA during workup for an unwitnessed fall. Her DKA was promptly managed, and she was subsequently diagnosed with metastatic pancreatic cancer. Given the advanced stage of her malignancy, the multidisciplinary team consensus was for a palliative approach. She passed away on day 10 of the admission. To our knowledge, this is the first report of a first DKA presentation as a manifestation of pancreatic cancer in an adult aged over 70 years. To date, there is no effective screening test for pancreatic cancer in the general population. However, new-onset DM in the appropriate context might indicate the need for further evaluation. While it is possible that unresectable tumours are identified, earlier diagnosis of DM with pancreatic cancer may facilitate more timely management, including earlier advanced care planning. Learning points A higher clinical suspicion for pancreatic cancer is required for older adults presenting with diabetic ketoacidosis without a previously diagnosed diabetes mellitus. A bi-directional relationship exists between diabetes and pancreatic cancer. Pancreatic cancer generally has a very poor prognosis due to its advanced stage at diagnosis and the lack of an effective screening test. New-onset diabetes in the appropriate context (such as weight loss) can indicate the need for further evaluation for underlying pancreatic cancer.
Collapse
Affiliation(s)
- Minh V Le
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Melbourne, Australia
| | - Spiros Fourlanos
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Australia
| | - Rahul D Barmanray
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
18
|
Varghese TS, Andrews C, Fisher L, Goldacre B, Mehrkar A, Pande R, Smith NAS, Walker AJ, Roberts KJ, Sultana A, MacKenna B, Lemanska A. Using Data to Improve Healthcare: A Case Study of Pancreatic Enzyme Replacement in Pancreatic Cancer. Semin Oncol Nurs 2024; 40:151688. [PMID: 39043534 DOI: 10.1016/j.soncn.2024.151688] [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: 02/03/2024] [Revised: 05/23/2024] [Accepted: 05/29/2024] [Indexed: 07/25/2024]
Abstract
OBJECTIVES In the UK, guidelines recommend pancreatic enzyme replacement therapy (PERT) to all people with unresectable pancreatic cancer. In 2023, we published a national audit of PERT which showed suboptimal prescribing and wide regional variation in England. The aim of this manuscript was to describe how we used the PERT audit to drive improvements in healthcare. METHODS Building on the PERT audit, we deployed an online dashboard which will deliver ongoing updates of the PERT audit. We developed a collaborative intervention with cancer nurse specialists (CNS) to improve care delivered to people with pancreatic cancer. The intervention called Creating a natiOnAL CNS pancrEatic cancer network to Standardise and improve CarE (COALESCE) will use the dashboard to evaluate improvements in prescribing of PERT. RESULTS We demonstrated how large databases of electronic healthcare records (EHRs) can be used to improve cancer care. The PERT audit was implemented into a dashboard for tracking the progress of COALESCE. We will measure improvements in PERT prescribing as the intervention with CNS progresses. CONCLUSIONS Improving healthcare is an ongoing and iterative process. By implementing the PERT dashboard, we created a resource-efficient, automated evaluation method enabling COALESCE to deliver a sustainable change. National-scale databases of EHRs enable rapid cycles of audits, providing regular feedback to interventions, working systematically to deliver change. Here, the focus is on pancreatic cancer. However, this methodology is transferable to other areas of healthcare. IMPLICATIONS FOR NURSING PRACTICE Nurses play a key role in collecting good quality data which are needed in clinical audits to identify shortcomings in healthcare. Nurse-driven interventions can be designed to improve healthcare. In this study, we capitalize on the unique role of CNS coordinating care for every patient with cancer. COALESCE is the first national collaborative study which uses CNS as researchers and change agents.
Collapse
Affiliation(s)
- Teena S Varghese
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Colm Andrews
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Louis Fisher
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Ben Goldacre
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Amir Mehrkar
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Rupaly Pande
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Nadia A S Smith
- Data Science Department, National Physical Laboratory, Teddington, UK
| | - Alex J Walker
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Keith J Roberts
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Asma Sultana
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Brian MacKenna
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Agnieszka Lemanska
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK; Data Science Department, National Physical Laboratory, Teddington, UK.
| |
Collapse
|
19
|
Grewe S, Jördens MS, Roderburg C, Leyh C, Labuhn S, Luedde T, Krieg S, Krieg A, Loosen SH, Kostev K. Elevated HbA1c Levels Are Associated with a Risk of Pancreatic Cancer: A Case-Control Study. J Clin Med 2024; 13:5584. [PMID: 39337070 PMCID: PMC11432739 DOI: 10.3390/jcm13185584] [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: 08/04/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Background: The early diagnosis of pancreatic cancer (ICD-10 C25) can improve the patient's prognosis. The association between pancreatic cancer and type 2 diabetes (T2D) is known, but not yet fully understood. It is, therefore, necessary to investigate the impact of hemoglobin A1c (HbA1c) serum levels on pancreatic cancer development and the potential intervention options. Methods: In the case-control study, patients from the German IQVIATM Disease Analyzer database aged ≥18 years with a diagnosis of pancreatic cancer (ICD-10 C25) and a diagnosis of T2D (ICD-10: E11) were included. The patients' propensity score matched 1:5 with individuals without pancreatic cancer. Logistic regression models were used to estimate odds ratios (ORs) with 95% confidence intervals (95% CI). Results: An elevated serum HbA1c prior to the index date was found to be significantly associated with an increased risk of a subsequent pancreatic cancer diagnosis for the mean HbA1c values of 6.5-8.4% (OR: 1.38; 95% CI: 1.22-1.57) as well as for mean HbA1c values ≥8.5% (OR: 1.41; 95% CI: 1.16-1.73). The only antihyperglycemic agent negatively associated with the subsequent pancreatic cancer diagnosis was the sodium-glucose cotransporter-2 (SGLT-2) inhibitor, with an odds ratio of 0.80 (95% confidence interval: 0.74-0.87 per year of therapy). This correlation was observed in both age- and sex-stratified subgroups. Conclusions: The data indicate that elevated serum HbA1c levels in patients with T2D are associated with an increased risk of pancreatic cancer development. It is possible that SGLT2 therapy may prove an effective means of reducing the risk of pancreatic cancer, thereby offering a potential avenue for the future reduction in pancreatic cancer incidence in patients with T2D.
Collapse
Affiliation(s)
- Steven Grewe
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Markus S Jördens
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Christoph Roderburg
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Catherine Leyh
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Simon Labuhn
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Sarah Krieg
- Department of Inclusive Medicine, University Hospital Ostwestfalen-Lippe, Bielefeld University, 33617 Bielefeld, Germany
| | - Andreas Krieg
- Department of General and Visceral Surgery, Thoracic Surgery and Proctology, University Hospital Herford, Medical Campus OWL, Ruhr University Bochum, 32049 Herford, Germany
| | - Sven H Loosen
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | | |
Collapse
|
20
|
Swain S, Narayan RK, Mishra PR. Unraveling the interplay: exploring signaling pathways in pancreatic cancer in the context of pancreatic embryogenesis. Front Cell Dev Biol 2024; 12:1461278. [PMID: 39239563 PMCID: PMC11374643 DOI: 10.3389/fcell.2024.1461278] [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: 07/08/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Pancreatic cancer continues to be a deadly disease because of its delayed diagnosis and aggressive tumor biology. Oncogenes and risk factors are being reported to influence the signaling pathways involved in pancreatic embryogenesis leading to pancreatic cancer genesis. Although studies using rodent models have yielded insightful information, the scarcity of human pancreatic tissue has made it difficult to comprehend how the human pancreas develops. Transcription factors like IPF1/PDX1, HLXB9, PBX1, MEIS, Islet-1, and signaling pathways, including Hedgehog, TGF-β, and Notch, are directing pancreatic organogenesis. Any derangements in the above pathways may lead to pancreatic cancer. TP53: and CDKN2A are tumor suppressor genes, and the mutations in TP53 and somatic loss of CDKN2A are the drivers of pancreatic cancer. This review clarifies the complex signaling mechanism involved in pancreatic cancer, the same signaling pathways in pancreas development, the current therapeutic approach targeting signaling molecules, and the mechanism of action of risk factors in promoting pancreatic cancer.
Collapse
Affiliation(s)
- Sashikanta Swain
- Department of Anatomy, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Ravi Kant Narayan
- Department of Anatomy, All India Institute of Medical Sciences, Bhubaneswar, India
| | | |
Collapse
|
21
|
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
|
22
|
Wang L, Levinson R, Mezzacappa C, Katona BW. Review of the cost-effectiveness of surveillance for hereditary pancreatic cancer. Fam Cancer 2024; 23:351-360. [PMID: 38795221 PMCID: PMC11255025 DOI: 10.1007/s10689-024-00392-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/16/2024] [Indexed: 05/27/2024]
Abstract
Individuals with hereditary pancreatic cancer risk include high risk individuals (HRIs) with germline genetic susceptibility to pancreatic cancer (PC) and/or a strong family history of PC. Previously, studies have shown that PC surveillance in HRIs can downstage PC diagnosis and extend survival leading to pancreatic surveillance being recommended for certain HRIs. However, the optimal surveillance strategy remains uncertain, including which modalities should be used for surveillance, how frequently should surveillance be performed, and which sub-groups of HRIs should undergo surveillance. Additionally, in the ideal world PC surveillance should also be cost-effective. Cost-effectiveness analysis is a valuable tool that can consider the costs, potential health benefits, and risks among various PC surveillance strategies. In this review, we summarize the cost-effectiveness of various PC surveillance strategies for HRIs for hereditary pancreatic cancer and provide potential avenues for future work in this field. Additionally, we include cost-effectiveness studies among individuals with new-onset diabetes (NoD), a high-risk group for sporadic PC, as a comparison.
Collapse
Affiliation(s)
- Louise Wang
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd. 751 South Pavilion, Philadelphia, PA, 19104, USA
| | - Rachel Levinson
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT, USA
| | | | - Bryson W Katona
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd. 751 South Pavilion, Philadelphia, PA, 19104, USA.
| |
Collapse
|
23
|
Khan S, Bhushan B. Machine Learning Predicts Patients With New-onset Diabetes at Risk of Pancreatic Cancer. J Clin Gastroenterol 2024; 58:681-691. [PMID: 37522752 DOI: 10.1097/mcg.0000000000001897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/22/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND New-onset diabetes represent a high-risk cohort to screen for pancreatic cancer. GOALS Develop a machine model to predict pancreatic cancer among patients with new-onset diabetes. STUDY A retrospective cohort of patients with new-onset diabetes was assembled from multiple health care networks in the United States. An XGBoost machine learning model was designed from a portion of this cohort (the training set) and tested on the remaining part of the cohort (the test set). Shapley values were used to explain the XGBoost's model features. Model performance was compared with 2 contemporary models designed to predict pancreatic cancer among patients with new-onset diabetes. RESULTS In the test set, the XGBoost model had an area under the curve of 0.80 (0.76 to 0.85) compared with 0.63 and 0.68 for other models. Using cutoffs based on the Youden index, the sensitivity of the XGBoost model was 75%, the specificity was 70%, the accuracy was 70%, the positive predictive value was 1.2%, and the negative predictive value was >99%. The XGBoost model obtained a positive predictive value of at least 2.5% with a sensitivity of 38%. The XGBoost model was the only model that detected at least 50% of patients with cancer one year after the onset of diabetes. All 3 models had similar features that predicted pancreatic cancer, including older age, weight loss, and the rapid destabilization of glucose homeostasis. CONCLUSION Machine learning models isolate a high-risk cohort from those with new-onset diabetes at risk for pancreatic cancer.
Collapse
Affiliation(s)
- Salman Khan
- Department of Medicine, West Virginia University School of Medicine, West Virginia University, Morgantown, WV
- Northeast Ohio Medical University, Rootstown, OH
| | - Bharath Bhushan
- Department of Medicine, West Virginia University School of Medicine, West Virginia University, Morgantown, WV
| |
Collapse
|
24
|
Jacobs MF, Stoffel EM. Genetic and other risk factors for pancreatic ductal adenocarcinoma (PDAC). Fam Cancer 2024; 23:221-232. [PMID: 38573398 DOI: 10.1007/s10689-024-00372-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 04/05/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is often diagnosed at an advanced stage, resulting in poor prognosis and low 5-year survival rates. While early evidence suggests increased long-term survival in those with screen-detected resectable cancers, surveillance imaging is currently only recommended for individuals with a lifetime risk of PDAC ≥ 5%. Identification of risk factors for PDAC provides opportunities for early detection, risk reducing interventions, and targeted therapies, thus potentially improving patient outcomes. Here, we summarize modifiable and non-modifiable risk factors for PDAC. We review hereditary cancer syndromes associated with risk for PDAC and their implications for patients and their relatives. In addition, other biologically relevant pathways and environmental and lifestyle risk factors are discussed. Future work may focus on elucidating additional genetic, environmental, and lifestyle risk factors that may modify PDAC risk to continue to identify individuals at increased risk for PDAC who may benefit from surveillance and risk reducing interventions.
Collapse
Affiliation(s)
- Michelle F Jacobs
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Elena M Stoffel
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| |
Collapse
|
25
|
Tan MCB, Isom CA, Liu Y, Trégouët DA, Wu L, Zhou D, Gamazon ER. Transcriptome-wide association study and Mendelian randomization in pancreatic cancer identifies susceptibility genes and causal relationships with type 2 diabetes and venous thromboembolism. EBioMedicine 2024; 106:105233. [PMID: 39002386 PMCID: PMC11284564 DOI: 10.1016/j.ebiom.2024.105233] [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: 01/18/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND Two important questions regarding the genetics of pancreatic adenocarcinoma (PDAC) are 1. Which germline genetic variants influence the incidence of this cancer; and 2. Whether PDAC causally predisposes to associated non-malignant phenotypes, such as type 2 diabetes (T2D) and venous thromboembolism (VTE). METHODS In this study of 8803 patients with PDAC and 67,523 controls, we first performed a large-scale transcriptome-wide association study to investigate the association between genetically determined gene expression in normal pancreas tissue and PDAC risk. Secondly, we used Mendelian Randomization (MR) to analyse the causal relationships among PDAC, T2D (74,124 cases and 824,006 controls) and VTE (30,234 cases and 172,122 controls). FINDINGS Sixteen genes showed an association with PDAC risk (FDR <0.10), including six genes not yet reported for PDAC risk (PPIP5K2, TFR2, HNF4G, LRRC10B, PRC1 and FBXL20) and ten previously reported genes (INHBA, SMC2, ABO, PDX1, MTMR6, ACOT2, PGAP3, STARD3, GSDMB, ADAM33). MR provided support for a causal effect of PDAC on T2D using genetic instruments in the HNF4G and PDX1 loci, and unidirectional causality of VTE on PDAC involving the ABO locus (OR 2.12, P < 1e-7). No evidence of a causal effect of PDAC on VTE was found. INTERPRETATION These analyses identified candidate susceptibility genes and disease relationships for PDAC that warrant further investigation. HNF4G and PDX1 may induce PDAC-associated diabetes, whereas ABO may induce the causative effect of VTE on PDAC. FUNDING National Institutes of Health (USA).
Collapse
Affiliation(s)
- Marcus C B Tan
- Division of Surgical Oncology and Endocrine Surgery, Section of Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Chelsea A Isom
- Herbert Wertheim School of Public Health & Human Longevity Science, University of California, San Diego, San Diego, CA, USA
| | - Yangzi Liu
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Lang Wu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Dan Zhou
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China.
| | - Eric R Gamazon
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Clare Hall, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
26
|
Goggins M. The role of biomarkers in the early detection of pancreatic cancer. Fam Cancer 2024; 23:309-322. [PMID: 38662265 PMCID: PMC11309746 DOI: 10.1007/s10689-024-00381-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 03/19/2024] [Indexed: 04/26/2024]
Abstract
Pancreatic surveillance can detect early-stage pancreatic cancer and achieve long-term survival, but currently involves annual endoscopic ultrasound and MRI/MRCP, and is recommended only for individuals who meet familial/genetic risk criteria. To improve upon current approaches to pancreatic cancer early detection and to expand access, more accurate, inexpensive, and safe biomarkers are needed, but finding them has remained elusive. Newer approaches to early detection, such as using gene tests to personalize biomarker interpretation, and the increasing application of artificial intelligence approaches to integrate complex biomarker data, offer promise that clinically useful biomarkers for early pancreatic cancer detection are on the horizon.
Collapse
Affiliation(s)
- Michael Goggins
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD, 21231, USA.
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
27
|
Yang J, Tan C, Liu Y, Zheng Z, Liu X, Chen Y. Remnant Pancreas Volume Affects New-Onset Impaired Glucose Homeostasis Secondary to Pancreatic Cancer. Biomedicines 2024; 12:1653. [PMID: 39200119 PMCID: PMC11351567 DOI: 10.3390/biomedicines12081653] [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: 06/21/2024] [Revised: 07/15/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND New-onset diabetes (NOD) has been identified as a high-risk factor for the early detection of pancreatic ductal adenocarcinoma (PDAC). The role of tumor volume and remnant pancreas volume (RPV) in the progression from normal to NOD in PDAC patients is not fully illustrated yet. METHODS In this cross-sectional study, glycemic metabolism traits of 95 PDAC patients before pancreatic surgery were described and compared with chronic pancreatitis and type 2 diabetes mellitus patients based on the oral glucose tolerance test. The remnant RPV and tumor volume, calculated by three-dimensional reconstruction of radiological images, were included in the ordinal logistic regression models. RESULTS The prevalence of NOD was high among PDAC patients (38.9%). However, normal glucose tolerance (NGT) or prediabetes mellitus status were present as more than half (24/44) of advanced tumor stage patients. Indexes reflecting beta-cell function but not insulin sensitivity gradually worsened from NGT to NOD patients (all p < 0.05). The remnant pancreas volume (RPV) was identified as a potential protective factor for diabetes secondary to PDAC (odds ratio 0.95, 95% CI [0.92, 0.97], p < 0.001). CONCLUSIONS Reduced RPV causing beta-cell dysfunction might be one of the mechanisms of NOD secondary to PDAC. Subjects with sufficient pancreas volume could not be detected earlier when regarding patients with NOD as the population at risk for PDAC.
Collapse
Affiliation(s)
- Jie Yang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu 610065, China
| | - Chunlu Tan
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ya Liu
- Department of Thyroid and Breast Surgery, Chengdu Second People’s Hospital, Chengdu 610041, China
| | - Zhenjiang Zheng
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xubao Liu
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yonghua Chen
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
28
|
Pliszka M, Szablewski L. Associations between Diabetes Mellitus and Selected Cancers. Int J Mol Sci 2024; 25:7476. [PMID: 39000583 PMCID: PMC11242587 DOI: 10.3390/ijms25137476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Cancer is one of the major causes of mortality and is the second leading cause of death. Diabetes mellitus is a serious and growing problem worldwide, and its prevalence continues to grow; it is the 12th leading cause of death. An association between diabetes mellitus and cancer has been suggested for more than 100 years. Diabetes is a common disease diagnosed among patients with cancer, and evidence indicates that approximately 8-18% of patients with cancer have diabetes, with investigations suggesting an association between diabetes and some particular cancers, increasing the risk for developing cancers such as pancreatic, liver, colon, breast, stomach, and a few others. Breast and colorectal cancers have increased from 20% to 30% and there is a 97% increased risk of intrahepatic cholangiocarcinoma or endometrial cancer. On the other hand, a number of cancers and cancer therapies increase the risk of diabetes mellitus. Complications due to diabetes in patients with cancer may influence the choice of cancer therapy. Unfortunately, the mechanisms of the associations between diabetes mellitus and cancer are still unknown. The aim of this review is to summarize the association of diabetes mellitus with selected cancers and update the evidence on the underlying mechanisms of this association.
Collapse
Affiliation(s)
- Monika Pliszka
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| | - Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| |
Collapse
|
29
|
Cosmin Stan M, Paul D. Diabetes and Cancer: A Twisted Bond. Oncol Rev 2024; 18:1354549. [PMID: 38835644 PMCID: PMC11148650 DOI: 10.3389/or.2024.1354549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 04/08/2024] [Indexed: 06/06/2024] Open
Abstract
This paper presents an overview of the interconnection between various factors related to both cancer and type 2 diabetes mellitus (T2DM). Hyperglycemia, hyperinsulinemia, chronic inflammation, and obesity are involved in the development and progression of both diseases but, strong evidence for a direct causal relationship between diabetes and cancer, is lacking. Several studies described a relationship between hyperglycemia and cancer at the cellular, tissular and organismic levels but at the same time recent Mendelian randomization studies proved a significant causal relationship only between hyperglycemia and breast cancer. On the other hand, the association between both hyperinsulinemia and obesity and several cancer types appears to be robust as demonstrated by Mendelian randomized studies. Metabolic alterations, including the Warburg effect and excessive glucose consumption by tumors, are discussed, highlighting the potential impact of dietary restrictions, such as fasting and low-carb diets, on tumor growth and inflammation. Recent data indicates that circulating branched-chain amino acids levels, may represent novel biomarkers that may contribute to both better diabetes control and early pancreatic cancer detection. Understanding the underlying mechanisms and shared risk factors between cancer and T2DM can provide valuable insights for cancer prevention, early detection, and management strategies.
Collapse
Affiliation(s)
- Mihai Cosmin Stan
- Emergency County Hospital Rm. Vâlcea, Râmnicu Vâlcea, Romania
- Medical Oncology Department, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Doru Paul
- Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
30
|
Wayne CD, Benbetka C, Besner GE, Narayanan S. Challenges of Managing Type 3c Diabetes in the Context of Pancreatic Resection, Cancer and Trauma. J Clin Med 2024; 13:2993. [PMID: 38792534 PMCID: PMC11122338 DOI: 10.3390/jcm13102993] [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: 04/10/2024] [Revised: 05/04/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Type 3c diabetes mellitus (T3cDM), also known as pancreatogenic or pancreoprivic diabetes, is a specific type of DM that often develops as a result of diseases affecting the exocrine pancreas, exhibiting an array of hormonal and metabolic characteristics. Several pancreatic exocrine diseases and surgical procedures may cause T3cDM. Diagnosing T3cDM remains difficult as the disease characteristics frequently overlap with clinical presentations of type 1 DM (T1DM) or type 2 DM (T2DM). Managing T3cDM is likewise challenging due to numerous confounding metabolic dysfunctions, including pancreatic endocrine and exocrine insufficiencies and poor nutritional status. Treatment of pancreatic exocrine insufficiency is of paramount importance when managing patients with T3cDM. This review aims to consolidate the latest information on surgical etiologies of T3cDM, focusing on partial pancreatic resections, total pancreatectomy, pancreatic cancer and trauma.
Collapse
Affiliation(s)
- Colton D. Wayne
- Department of Pediatric Surgery, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA; (C.D.W.); (G.E.B.)
- Center for Perinatal Research, Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Surgery, Baylor University Medical Center, 3600 Gaston Ave, Dallas, TX 75246, USA
| | | | - Gail E. Besner
- Department of Pediatric Surgery, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA; (C.D.W.); (G.E.B.)
- Center for Perinatal Research, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Siddharth Narayanan
- Department of Pediatric Surgery, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA; (C.D.W.); (G.E.B.)
- Center for Perinatal Research, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| |
Collapse
|
31
|
Khayat S, Choudhary K, Claude Nshimiyimana J, Gurav J, Hneini A, Nazir A, Chaito H, Wojtara M, Uwishema O. Pancreatic cancer: from early detection to personalized treatment approaches. Ann Med Surg (Lond) 2024; 86:2866-2872. [PMID: 38694319 PMCID: PMC11060269 DOI: 10.1097/ms9.0000000000002011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 03/19/2024] [Indexed: 05/04/2024] Open
Abstract
Pancreatic cancer is notorious for its persistently poor prognosis and health outcomes, so some of the questions that may be begged are "Why is it mostly diagnosed at end stage?", "What could we possibly do with the advancing technology in today's world to detect early pancreatic cancer and intervene?", and "Are there any implementation of the existing novel imaging technologies?". Well, to start with, this is in part because the majority of patients presented would already have reached a locally advanced or metastatic stage at the time of diagnosis due to its highly aggressive characteristics and lack of symptoms. Due to this striking disparity in survival, advancements in early detection and intervention are likely to significantly increase patients' survival. Presently, screening is frequently used in high-risk individuals in order to obtain an early pancreatic cancer diagnosis. Having a thorough understanding of the pathogenesis and risk factors of pancreatic cancer may enable us to identify individuals at high risk, diagnose the disease early, and begin treatment promptly. In this review, the authors outline the clinical hurdles to early pancreatic cancer detection, describe high-risk populations, and discuss current screening initiatives for high-risk individuals. The ultimate goal of this current review is to study the roles of both traditional and novel imaging modalities for early pancreatic cancer detection. A lot of the novel imaging techniques mentioned seem promising, but they need to be put to the test on a large scale and may need to be combined with other non-invasive biomarkers before they can be widely used.
Collapse
Affiliation(s)
| | | | | | | | - Asmaa Hneini
- Faculty of Medicine, Beirut Arab University, Beirut, Lebanon
| | - Abubakar Nazir
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Hassan Chaito
- Faculty of Medicine, Beirut Arab University, Beirut, Lebanon
| | - Magda Wojtara
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
| | - Olivier Uwishema
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
32
|
Borgmästars E, Jacobson S, Simm M, Johansson M, Billing O, Lundin C, Nyström H, Öhlund D, Lubovac-Pilav Z, Jonsson P, Franklin O, Sund M. Metabolomics for early pancreatic cancer detection in plasma samples from a Swedish prospective population-based biobank. J Gastrointest Oncol 2024; 15:755-767. [PMID: 38756646 PMCID: PMC11094504 DOI: 10.21037/jgo-23-930] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/31/2024] [Indexed: 05/18/2024] Open
Abstract
Background Pancreatic ductal adenocarcinoma (pancreatic cancer) is often detected at late stages resulting in poor overall survival. To improve survival, more patients need to be diagnosed early when curative surgery is feasible. We aimed to identify circulating metabolites that could be used as early pancreatic cancer biomarkers. Methods We performed metabolomics by liquid and gas chromatography-mass spectrometry in plasma samples from 82 future pancreatic cancer patients and 82 matched healthy controls within the Northern Sweden Health and Disease Study (NSHDS). Logistic regression was used to assess univariate associations between metabolites and pancreatic cancer risk. Least absolute shrinkage and selection operator (LASSO) logistic regression was used to design a metabolite-based risk score. We used receiver operating characteristic (ROC) analyses to assess the discriminative performance of the metabolite-based risk score. Results Among twelve risk-associated metabolites with a nominal P value <0.05, we defined a risk score of three metabolites [indoleacetate, 3-hydroxydecanoate (10:0-OH), and retention index (RI): 2,745.4] using LASSO. A logistic regression model containing these three metabolites, age, sex, body mass index (BMI), smoking status, sample date, fasting status, and carbohydrate antigen 19-9 (CA 19-9) yielded an internal area under curve (AUC) of 0.784 [95% confidence interval (CI): 0.714-0.854] compared to 0.681 (95% CI: 0.597-0.764) for a model without these metabolites (P value =0.007). Seventeen metabolites were significantly associated with pancreatic cancer survival [false discovery rate (FDR) <0.1]. Conclusions Indoleacetate, 3-hydroxydecanoate (10:0-OH), and RI: 2,745.4 were identified as the top candidate biomarkers for early detection. However, continued efforts are warranted to determine the usefulness of these metabolites as early pancreatic cancer biomarkers.
Collapse
Affiliation(s)
- Emmy Borgmästars
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
| | - Sara Jacobson
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
| | - Maja Simm
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
- Department of Clinical Sciences/Obstetrics and Gynecology, Umeå University, Umeå, Sweden
| | - Mattias Johansson
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Ola Billing
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
| | - Christina Lundin
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
| | - Hanna Nyström
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Daniel Öhlund
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
- Department of Radiation Sciences/Oncology, Umeå University, Umeå, Sweden
| | | | - Pär Jonsson
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Oskar Franklin
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, USA
| | - Malin Sund
- Department of Surgical and Perioperative Sciences/Surgery, Umeå University, Umeå, Sweden
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
33
|
Conway RB, Hudson AG, Munro H, Fu D, McClain DA, Blot WJ. Diabetes and pancreatic cancer risk in a multiracial cohort. Diabet Med 2024; 41:e15234. [PMID: 37779225 PMCID: PMC11357321 DOI: 10.1111/dme.15234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/03/2023]
Abstract
AIMS To determine the relationship of diabetes with pancreatic cancer incidence among African American and Whites of similar socio-economic status. METHODS Using the Southern Community Cohort Study, we conducted a follow-up during 2002-2015 of pancreatic cancer incidence of 73,378 mostly low-income participants aged 40-79 years; 15,913 reported diabetes at baseline. Multivariable Cox analysis controlling for sex, family history of pancreatic cancer, BMI, smoking status, alcohol consumption, education, income and other important covariates, and with age as the timescale was used. RESULTS Totally, 265 incident pancreatic cancer cases were observed. Pancreatic cancer risk was increased among those with diabetes (HR 1.54, CI 1.16-2.05), with similar increases among African Americans (HR 1.51, CI 1.08-2.11) and Whites (HR 1.78, CI 1.00-3.16). No trend in risk was observed for diabetes duration among those with diabetes, with HRs of 1.39 (0.91-2.11), 2.31 (1.51-3.54) and 1.23 (0.80-1.89) for <5, 5-9 and 10+ years duration, respectively. African Americans were at increased risk of pancreatic cancer (HR = 1.40, 95% CI 1.05-1.87), which persisted after adjusting for diabetes (HR 1.36, CI 1.02-1.81). The effect sizes for other pancreatic cancer risk factors with pancreatic cancer were similar by diabetes status, although a stronger association with low BMI was evident among those with diabetes. CONCLUSIONS Diabetes increases pancreatic cancer risk similarly among African Americans and Whites in this Southern U.S. COHORT
Collapse
Affiliation(s)
- Rebecca B.N. Conway
- American Academy of Epidemiology, Inc., 2008 S. Wiley Avenue, Tyler, TX 75701, USA
- Department of Epidemiology, University of Colorado School of Public Health, Anschutz Medical Campus, 13001 East 17th Place, Mail Stop B119 Aurora, CO, 80045, USA
| | - Alana G. Hudson
- Public Health Strategies, LLC, 515 Highland Avenue, South Charleston, WV, 25303, USA
| | - Heather Munro
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, 2525 West End Avenue Nashville, TN, 37203
| | - David Fu
- Cancer Center, Renmin Hospital of Wuhan University, No. 99 Zhangzhidong St., Wuchang District, Wuhan City, Hubei Province, 430060, P.R. China
| | - Donald A. McClain
- Section of Endocrinology and Metabolism, Wake Forest School of Medicine, 475 Vine St, Winston-Salem, NC, 27157, USA
| | - William J. Blot
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, 2525 West Avenue, Nashville, TN, 27203, USA
| |
Collapse
|
34
|
Wang Z, Zhang G, Fu J, Li G, Zhao Z, Choe H, Ding K, Ma J, Wei J, Shang D, Zhang L. Mechanism exploration and biomarker identification of glycemic deterioration in patients with diseases of the exocrine pancreas. Sci Rep 2024; 14:4374. [PMID: 38388766 PMCID: PMC10883946 DOI: 10.1038/s41598-024-52956-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
The damage to the endocrine pancreas among patients with diseases of the exocrine pancreas (DP) leads to reduced glycemic deterioration, ultimately resulting in diabetes of the exocrine pancreas (DEP). The present research aims to investigate the mechanism responsible for glycemic deterioration in DP patients, and to identify useful biomarkers, with the ultimate goal of enhancing clinical practice awareness. Gene expression profiles of patients with DP in this study were acquired from the Gene Expression Omnibus database. The original study defines DP patients to belong in one of three categories: non-diabetic (ND), impaired glucose tolerance (IGT) and DEP, which correspond to normoglycemia, early and late glycemic deterioration, respectively. After ensuring quality control, the discovery cohort included 8 ND, 20 IGT, and 12 DEP, while the validation cohort included 27 ND, 15 IGT, and 20 DEP. Gene set enrichment analysis (GSEA) employed differentially expressed genes (DEGs), while immunocyte infiltration was determined using single sample gene set enrichment analysis (ssGSEA). Additionally, correlation analysis was conducted to establish the link between clinical characteristics and immunocyte infiltration. The least absolute shrinkage and selection operator regression and random forest combined to identify biomarkers indicating glycemic deterioration in DP patients. These biomarkers were further validated through independent cohorts and animal experiments. With glycemic deterioration, biological processes in the pancreatic islets such as nutrient metabolism and complex immune responses are disrupted in DP patients. The expression of ACOT4, B2M, and ACKR2 was upregulated, whereas the expression of CACNA1F was downregulated. Immunocyte infiltration in the islet microenvironment showed a significant positive correlation with the age, body mass index (BMI), HbA1c and glycemia at the 2-h of patients. It was a crucial factor in glycemic deterioration. Additionally, B2M demonstrated a significant positive correlation with immunocyte infiltration and clinical features. Quantitative real-time PCR (qRT-PCR) and western blotting confirmed the upregulation in B2M. Immunofluorescent staining suggested the alteration of B2M was mainly in the alpha cells and beta cells. Overall, the study showed that gradually increased immunocyte infiltration was a significant contributor to glycemic deterioration in patients with DP, and it also highlighted B2M as a biomarker.
Collapse
Affiliation(s)
- Zhen Wang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Guolin Zhang
- Department of Cardiology II, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Jixian Fu
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Guangxing Li
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Zhihao Zhao
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - HyokChol Choe
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
- Department of Clinical Medicine, Sinuiju Medical University, Sinuiju, Republic of Korea
| | - Kaiyue Ding
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Junnan Ma
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Jing Wei
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China.
| | - Dong Shang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China.
| | - Lin Zhang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
35
|
Cohrs CM, Chen C, Atkinson MA, Drotar DM, Speier S. Bridging the Gap: Pancreas Tissue Slices From Organ and Tissue Donors for the Study of Diabetes Pathogenesis. Diabetes 2024; 73:11-22. [PMID: 38117999 PMCID: PMC10784654 DOI: 10.2337/dbi20-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 10/14/2023] [Indexed: 12/22/2023]
Abstract
Over the last two decades, increased availability of human pancreatic tissues has allowed for major expansions in our understanding of islet biology in health and disease. Indeed, studies of fixed and frozen pancreatic tissues, as well as efforts using viable isolated islets obtained from organ donors, have provided significant insights toward our understanding of diabetes. However, the procedures associated with islet isolation result in distressed cells that have been removed from any surrounding influence. The pancreas tissue slice technology was developed as an in situ approach to overcome certain limitations associated with studies on isolated islets or fixed tissue. In this Perspective, we discuss the value of this novel platform and review how pancreas tissue slices, within a short time, have been integrated in numerous studies of rodent and human islet research. We show that pancreas tissue slices allow for investigations in a less perturbed organ tissue environment, ranging from cellular processes, over peri-islet modulations, to tissue interactions. Finally, we discuss the considerations and limitations of this technology in its future applications. We believe the pancreas tissue slices will help bridge the gap between studies on isolated islets and cells to the systemic conditions by providing new insight into physiological and pathophysiological processes at the organ level. ARTICLE HIGHLIGHTS Human pancreas tissue slices represent a novel platform to study human islet biology in close to physiological conditions. Complementary to established technologies, such as isolated islets, single cells, and histological sections, pancreas tissue slices help bridge our understanding of islet physiology and pathophysiology from single cell to intact organ. Diverse sources of viable human pancreas tissue, each with distinct characteristics to be considered, are available to use in tissue slices for the study of diabetes pathogenesis.
Collapse
Affiliation(s)
- Christian M. Cohrs
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Chunguang Chen
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Mark A. Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL
| | - Denise M. Drotar
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL
| | - Stephan Speier
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Munich at the University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
36
|
Kiritani S, Ono Y, Takamatsu M, Yoshio S, Miyashita M, Oba A, Sato T, Ito H, Inoue Y, Saiura A, Takahashi Y. Unique Biology of Pancreatic Ductal Adenocarcinoma Accompanied by Rapidly Impaired Diabetes: A Favorable Long-Term Survival Following Curative Resection. Ann Surg Oncol 2024; 31:514-524. [PMID: 37803089 DOI: 10.1245/s10434-023-14408-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/17/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinomas (PDACs) are sometimes diagnosed accompanied by rapidly impaired diabetes (PDAC-RID). Although this type of PDAC may have unusual biological features, these features have not been explained. METHODS Patients with PDAC who underwent upfront pancreatectomy between 2010 and 2018 were retrospectively reviewed. PDAC-RID was defined as a glycated hemoglobin (HbA1c) value of ≥ 8.0% of newly diagnosed diabetes, and acute exacerbation of previously diagnosed diabetes. Other patients were classified as PDAC with stable glycometabolism (PDAC-SG). Clinicopathological factors, long-term survival rates, and recurrence patterns were evaluated. RESULTS Of the 520 enrolled patients, 104 were classified as PDAC-RID and 416 as PDAC-SG. There was no significant difference regarding TNM staging, resectability, or adjuvant chemotherapy rate between the groups. However, 5-years cancer-specific survival (CSS) was significantly higher in the PDAC-RID group than in the PDAC-SG group (45.3% vs. 31.1%; p = 0.02). This survival difference was highlighted in relatively early-stage PDAC (≤ pT2N1) (CSS: 60.8% vs. 43.6%; p = 0.01), but the difference was not significant for advanced-stage PDAC. A multivariate analysis of early-stage PDAC showed that PDAC-SG was an independent risk factor of shorter CSS (hazard ratio 1.76; p = 0.02). The hematogenous metastatic rate in early-stage PDAC was lower in the PDAC-RID group than in the PDAC-SG group (18.3% vs. 35.8%; p = 0.01). CONCLUSIONS PDAC-RID showed a favorable long-term survival rate after curative resection with low hematogenous metastases, which may be due to its unique biology.
Collapse
Affiliation(s)
- Sho Kiritani
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoshihiro Ono
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Manabu Takamatsu
- Department of Pathology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Sachiyo Yoshio
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Mamiko Miyashita
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Atsushi Oba
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takafumi Sato
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiromichi Ito
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yosuke Inoue
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akio Saiura
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Yu Takahashi
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan.
| |
Collapse
|
37
|
Yoo D, Kang M, Jung J. Risk of Ischemic Heart Disease in Patients With Postpancreatectomy Diabetes and Pancreatic Cancer: A Population-Based Study. J Am Heart Assoc 2023; 12:e031321. [PMID: 38084734 PMCID: PMC10863790 DOI: 10.1161/jaha.123.031321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 11/09/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Postpancreatectomy diabetes can be caused by resection of functioning pancreatic tissue and is associated with postoperative pancreatic islet cell mass loss and subsequent endocrine dysfunction. Diabetes is a well-known risk factor for ischemic heart disease. However, no previous studies have investigated ischemic heart disease in patients with postpancreatectomy diabetes and pancreatic cancer. METHODS AND RESULTS Rates of patients with diabetes diagnosed with pancreatic cancer who underwent pancreatectomy between 2002 and 2019 in South Korea were obtained from the Korean National Health Insurance Service database. Patient-level propensity score matching was conducted to reduce the possibility of selection bias, and multivariate Cox proportional hazards models were used to determine the association between postpancreatectomy diabetes and ischemic heart disease. In total, 30 242 patients were initially enrolled in the study. After applying exclusion criteria and propensity score matching, 2952 patients were included in the comparative analysis between the postpancreatectomy group with diabetes and the group without diabetes. Patients in the postpancreatectomy group with diabetes had significantly higher rates of ischemic heart disease than those in the group without diabetes. In total, 3432 patients were included in the comparison between the postpancreatectomy and prepancreatectomy groups with diabetes. There was no significant difference in the risk of ischemic heart disease between the postpancreatectomy and prepancreatectomy groups with diabetes. CONCLUSIONS Patients who developed diabetes after pancreatectomy had a higher risk of ischemic heart disease than patients who did not develop diabetes after pancreatectomy, and the rate of ischemic heart disease in these patients was similar to that in patients preoperatively diagnosed with diabetes.
Collapse
Affiliation(s)
- Daegwang Yoo
- Division of Hepatobiliary and Pancreatic Surgery, Department of SurgerySoonchunhyang University College of Medicine, Soonchunhyang University Seoul HospitalSeoulSouth Korea
| | - Minsun Kang
- Artificial Intelligence and Big‐Data Convergence Center, Gil Medical CenterGachon University College of MedicineIncheonSouth Korea
| | - Jaehun Jung
- Artificial Intelligence and Big‐Data Convergence Center, Gil Medical CenterGachon University College of MedicineIncheonSouth Korea
- Department of Preventive MedicineGachon University College of MedicineIncheonSouth Korea
| |
Collapse
|
38
|
Schranz A, Sternad C, Aziz F, Wagner D, Kornprat P, Sucher R, Jost PJ, Wölfler A, Pieber TR, Sourij H, Riedl JM, Aberer F. Incidence of Diabetes Mellitus and Its Impact on Outcomes in Patients Undergoing Surgical Pancreatectomy for Non-Malignant and Malignant Pancreatobiliary Diseases-A Retrospective Analysis. J Clin Med 2023; 12:7532. [PMID: 38137600 PMCID: PMC10744322 DOI: 10.3390/jcm12247532] [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: 10/27/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Diabetes mellitus (DM) is a prominent risk factor for malignant and non-malignant pancreatic diseases. Furthermore, the presence of DM predicts an unfavourable outcome in people with pancreatic cancer. This retrospective observational study investigated 370 patients who underwent pancreatic resection surgery for various indications (84.3% in malignant indication) in a single surgery centre in Graz, Austria. The preoperative and postoperative diabetes statuses were evaluated according to surgery method and disease entity and predictors for diabetes development after surgery, as well as outcomes (survival and cancer recurrence) according to diabetes status, were analysed. In the entire cohort, the postoperative diabetes (postopDM) incidence was 29%. PostopDM occurred significantly more frequently in malignoma patients than in those with benign diseases (31.3% vs. 16.7%; p = 0.040, OR = 2.28). In the malignoma population, BMI, longer surgery duration, and prolonged ICU and hospital stay were significant predictors of diabetes development. The 1- and 2-year follow-ups showed a significantly increased mortality of people with postopDM in comparison to people without diabetes (HR 1-year = 2.02, p = 0.014 and HR 2-years = 1.56, p = 0.034). Local cancer recurrence was not influenced by the diabetes status. Postoperative new-onset diabetes seems to be associated with higher mortality of patients with pancreatic malignoma undergoing pancreatobiliary surgery.
Collapse
Affiliation(s)
- Anna Schranz
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (A.S.); (C.S.); (F.A.); (T.R.P.); (F.A.)
| | - Christoph Sternad
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (A.S.); (C.S.); (F.A.); (T.R.P.); (F.A.)
| | - Faisal Aziz
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (A.S.); (C.S.); (F.A.); (T.R.P.); (F.A.)
| | - Doris Wagner
- Department of Surgery, Medical University of Graz, 8036 Graz, Austria; (D.W.); (P.K.); (R.S.)
| | - Peter Kornprat
- Department of Surgery, Medical University of Graz, 8036 Graz, Austria; (D.W.); (P.K.); (R.S.)
| | - Robert Sucher
- Department of Surgery, Medical University of Graz, 8036 Graz, Austria; (D.W.); (P.K.); (R.S.)
| | - Philipp J. Jost
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036 Graz, Austria; (P.J.J.); (J.M.R.)
| | - Albert Wölfler
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, 8036 Graz, Austria;
| | - Thomas R. Pieber
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (A.S.); (C.S.); (F.A.); (T.R.P.); (F.A.)
| | - Harald Sourij
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (A.S.); (C.S.); (F.A.); (T.R.P.); (F.A.)
| | - Jakob M. Riedl
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036 Graz, Austria; (P.J.J.); (J.M.R.)
| | - Felix Aberer
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (A.S.); (C.S.); (F.A.); (T.R.P.); (F.A.)
| |
Collapse
|
39
|
Apte M. A journey to and with the stars: The pancreatic stellate cell story. Pancreatology 2023; 23:893-899. [PMID: 37973449 DOI: 10.1016/j.pan.2023.10.024] [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: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
The George E Palade Prize is the highest honour awarded by the International Association of Pancreatology, that recognises an individual who has made outstanding contributions to the understanding of the pancreas and pancreatic diseases. The 2023 Palade Prize was awarded to Professor Minoti Apte, University of New South Wales Sydney on September 16, 2023 during the Joint Meeting of the International Association of Pancreatology and the Indian Pancreas Club, held in Delhi, India. This paper summarises her Palade lecture wherein she reflects on her journey as a medical graduate, an academic and a researcher, with a particular focus on her team's pioneering work on pancreatic stellate cell biology and the role of these cells in health and disease. While there has been much progress in this field with the efforts of researchers worldwide, there is much still to be learned; thus it is a topic with ample scope for innovative research with the potential to translate into better outcomes for patients with pancreatic disease.
Collapse
Affiliation(s)
- Minoti Apte
- Pancreatic Research Group, South Western Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney and Ingham Institute for Applied Medical Research, Liverpool, Sydney, Australia.
| |
Collapse
|
40
|
Abstract
Obesity has been recognized to be increasing globally and is designated a disease with adverse consequences requiring early detection and appropriate care. In addition to being related to metabolic syndrome disorders such as type 2 diabetes, hypertension, stroke, and premature coronary artery disease. Obesity is also etiologically linked to several cancers. The non-gastrointestinal cancers are breast, uterus, kidneys, ovaries, thyroid, meningioma, and thyroid. Gastrointestinal (GI) cancers are adenocarcinoma of the esophagus, liver, pancreas, gallbladder, and colorectal. The brighter side of the problem is that being overweight and obese and cigarette smoking are mostly preventable causes of cancers. Epidemiology and clinical studies have revealed that obesity is heterogeneous in clinical manifestations. In clinical practice, BMI is calculated by dividing a person's weight in kilograms by the square of the person's height in square meters (kg/m2). A BMI above 30 kg/m2 (defining obesity in many guidelines) is considered obesity. However, obesity is heterogeneous. There are subdivisions for obesity, and not all obesities are equally pathogenic. Adipose tissue, in particular, visceral adipose tissue (VAT), is endocrine and abdominal obesity (a surrogate for VAT) is evaluated by waist-hip measurements or just waist measures. Visceral Obesity, through several hormonal mechanisms, induces a low-grade chronic inflammatory state, insulin resistance, components of metabolic syndrome, and cancers. Metabolically obese, normal-weight (MONW) individuals in several Asian countries may have BMI below normal levels to diagnose obesity but suffer from many obesity-related complications. Conversely, some people have high BMI but are generally healthy with no features of metabolic syndrome. Many clinicians advise weight loss by dieting and exercise to metabolically healthy obese with large body habitus than to individuals with metabolic obesity but normal BMI. The GI cancers (esophagus, pancreas, gallbladder, liver, and colorectal) are individually discussed, emphasizing the incidence, possible pathogenesis, and preventive measures. From 2005 to 2014, most cancers associated with overweight and Obesity increased in the United States, while cancers related to other factors decreased. The standard recommendation is to offer or refer adults with a body mass index (BMI) of 30 or more to intensive, multicomponent behavioral interventions. However, the clinicians have to go beyond. They should critically evaluate BMI with due consideration for ethnicity, body habitus, and other factors that influence the type of obesity and obesity-related risks. In 2001, the Surgeon General's ``Call to Action to Prevent and Decrease Overweight and Obesity'' identified obesity as a critical public health priority for the United States. At government levels reducing obesity requires policy changes that improve the food and physical activity for all. However, implementing some policies with the most significant potential benefit to public health is politically tricky. The primary care physician, as well as subspecialists, should identify overweight and Obesity based on all the variable factors in the diagnosis. The medical community should address the prevention of overweight and Obesity as an essential part of medical care as much as vaccination in preventing infectious diseases at all levels- from childhood, to adolescence, and adults.
Collapse
Affiliation(s)
- Yuntao Zou
- Department of Medicine, Saint Peter's University Hospital, 125 Andover DR, Kendall Park, New Brunswick, NJ 08901, USA
| | - Capecomorin S Pitchumoni
- Department of Medicine, Saint Peter's University Hospital, 125 Andover DR, Kendall Park, New Brunswick, NJ 08901, USA.
| |
Collapse
|
41
|
Ekström E, Fagher K, Tingstedt B, Rystedt J, Nilsson J, Löndahl M, Andersson B. Hyperglycemia and insulin infusion in pancreatoduodenectomy: a prospective cohort study on feasibility and impact on complications. Int J Surg 2023; 109:3770-3777. [PMID: 37720940 PMCID: PMC10720831 DOI: 10.1097/js9.0000000000000714] [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: 04/13/2023] [Accepted: 08/13/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND Hyperglycemia is a risk factor for postoperative complications but its impact on outcome after pancreatoduodenectomy (PD) is scarcely studied. This prospective cohort study aimed to assess the effect of continuous insulin infusion on postoperative complications and blood glucose, as well as to evaluate the impact of hyperglycemia on complications, after PD. MATERIALS AND METHODS One hundred patients planned for PD at Skåne University Hospital, Sweden were prospectively included for perioperative continuous insulin infusion and a historic cohort of 100 patients was included retrospectively. Median blood glucose levels were calculated and data on complications were analyzed and compared between the historic cohort and the intervention group as well as between normo- and hyperglycemic patients. RESULTS Median glucose levels were significantly lower in the intervention group compared to the historic cohort up to 30 days postoperatively (median glucose 8.5 mmol/l (interquartile range 6.4-11) vs. 9.1 mmol/l (interquartile range 6.8-17) ( P =0.007)). No significant differences in complication rates were recorded between these two groups. The incidence of complications classified as Clavien ≥3 was higher in hyperglycemic patients (100 vs. 27%, P =0.024). Among hyperglycemic patients the prevalence of preoperative diabetes was higher compared to normoglycemic patients (52 vs.12%, P <0.001). In patients with a known diagnosis of diabetes, a trend, although not statistically significant, towards a lower incidence of postoperative pancreatic fistula grade B and C, as well as postpancreatectomy hemorrhage grade B and C, was seen compared to those without preoperative diabetes (6.8 vs. 14%, P =0.231 and 2.3 vs. 7.0%, P =0.238, respectively). CONCLUSION Insulin infusion in the early postoperative phase after PD is feasible in a non-ICU setting and significantly decreased blood glucose levels. The influence on complications was limited. Preoperative diabetes was a significant predictor of postoperative hyperglycemia and was associated with a lower incidence of clinically significant postoperative pancreatic fistula.
Collapse
Affiliation(s)
- Eva Ekström
- Department of Surgery, Skåne University Hospital
- Department of Clinical Sciences, Surgery, Lund University
| | - Katarina Fagher
- Department of Clinical Sciences, Surgery, Lund University
- Department of Endocrinology Skåne University Hospital
| | - Bobby Tingstedt
- Department of Surgery, Skåne University Hospital
- Department of Clinical Sciences, Surgery, Lund University
| | - Jenny Rystedt
- Department of Surgery, Skåne University Hospital
- Department of Clinical Sciences, Surgery, Lund University
| | - Johan Nilsson
- Department of Cardiothoracic Surgery, Skåne University Hospital
- Department of Translational Medicine, Lund University, Lund, Sweden
| | - Magnus Löndahl
- Department of Clinical Sciences, Surgery, Lund University
- Department of Endocrinology Skåne University Hospital
| | - Bodil Andersson
- Department of Surgery, Skåne University Hospital
- Department of Clinical Sciences, Surgery, Lund University
| |
Collapse
|
42
|
Senavirathna L, Pan S, Chen R. Protein Advanced Glycation End Products and Their Implications in Pancreatic Cancer. Cancer Prev Res (Phila) 2023; 16:601-610. [PMID: 37578815 PMCID: PMC10843555 DOI: 10.1158/1940-6207.capr-23-0162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/14/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
Protein advanced glycation end products (AGE) formed by nonenzymatic glycation can disrupt the normal structure and function of proteins, and stimulate the receptor for AGEs (RAGE), triggering intricate mechanisms that are etiologically related to various chronic diseases, including pancreatic cancer. Many common risk factors of pancreatic cancer are the major sources for the formation of protein AGEs and glycative stress in the human body. Abnormal accumulation of protein AGEs can impair the cellular proteome and promote AGE-RAGE driven pro-inflammatory signaling cascades, leading to increased oxidative stress, protease resistance, protein dysregulation, transcription activity of STAT, NF-κB, and AP-1, aberrant status in ubiquitin-proteasome system and autophagy, as well as other molecular events that are susceptible for the carcinogenic transformation towards the development of neoplasms. Here, we review studies to highlight our understanding in the orchestrated molecular events in bridging the impaired proteome, dysregulated functional networks, and cancer hallmarks initiated upon protein AGE formation and accumulation in pancreatic cancer.
Collapse
Affiliation(s)
- Lakmini Senavirathna
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Sheng Pan
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ru Chen
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
43
|
Wang L, Li X, Wu J, Tang Q. Pancreatic Cancer-Derived Exosomal miR-Let-7b-5p Stimulates Insulin Resistance in Skeletal Muscle Cells Through RNF20/STAT3/FOXO1 Axis Regulation. Diabetes Metab Syndr Obes 2023; 16:3133-3145. [PMID: 37842335 PMCID: PMC10573399 DOI: 10.2147/dmso.s430443] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/28/2023] [Indexed: 10/17/2023] Open
Abstract
Background Cancers trigger systemic metabolic disorders usually associated with glucose intolerance, which is an initially apparent phenomenon. One of the features of pancreatic cancer (PC) metabolic reprogramming is the crosstalk between PC and peripheral tissues (skeletal muscle and adipose tissues), emphasized by insulin resistance (IR). Our previous study reported that mice pancreatic cancer-derived exosomes could induce skeletal muscle cells (C2C12) IR, and exosomal microRNAs (miRNAs) may exert an important effect. However, the underlying mechanism remains to be further elucidated. Methods qPCR was used to determine the expression of let-7b-5p in normal pancreatic islet cells and PC cells. Exosomes were purified from PC cell culture medium by ultracentrifugation. The role let-7b-5p on IR-mediated by PC cells-derived exosomes was asses by Oil Red O staining using miRNA inhibitor. Western blot assay was performed to examine the expression of IR-related genes and the activation of signaling pathways. A Luciferase experiment was applied to confirm how let-7b-5p regulated the expression of RNF20. IP/WB analysis further determined whether RNF20 promoted STAT3 ubiquitination. Rescue experiment using RNF20 overexpression plasmid was performed to confirm the role of RNF20 on IR-mediated using PC cell-derived exosomes in C2C12 myotube cells. Results miRNA-let-7b-5p was identified as the key exosomal miRNA, which could promote the IR in C2C12 myotube cells supported the lipid accumulation, the activation of STAT3/FOXO1 axis, and the decreased expression of IRS-1 and GLUT4. RNF20, an E3 ubiquitin ligase, was confirmed as the target gene of let-7b-5p and was found to improve IR by downregulating STAT3 protein expression via ubiquitination-mediated protein degradation. The ectopic expression of RNF20 could effectively attenuate the IR mediated by the pancreatic cancer-derived exosomes in C2C12 myotube cells. Conclusion Our data suggest that exosomal miRNA-let-7b-5p may promote IR in C2C12 myotube cells by targeting RNF20 to activate the STAT3/FOXO1 axis.
Collapse
Affiliation(s)
- Lantian Wang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Xiawei Li
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Jian Wu
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Qiang Tang
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
44
|
Irajizad E, Kenney A, Tang T, Vykoukal J, Wu R, Murage E, Dennison JB, Sans M, Long JP, Loftus M, Chabot JA, Kluger MD, Kastrinos F, Brais L, Babic A, Jajoo K, Lee LS, Clancy TE, Ng K, Bullock A, Genkinger JM, Maitra A, Do KA, Yu B, Wolpin BM, Hanash S, Fahrmann JF. A blood-based metabolomic signature predictive of risk for pancreatic cancer. Cell Rep Med 2023; 4:101194. [PMID: 37729870 PMCID: PMC10518621 DOI: 10.1016/j.xcrm.2023.101194] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/20/2022] [Accepted: 08/21/2023] [Indexed: 09/22/2023]
Abstract
Emerging evidence implicates microbiome involvement in the development of pancreatic cancer (PaCa). Here, we investigate whether increases in circulating microbial-related metabolites associate with PaCa risk by applying metabolomics profiling to 172 sera collected within 5 years prior to PaCa diagnosis and 863 matched non-subject sera from participants in the Prostate, Lung, Colorectal, and Ovarian (PLCO) cohort. We develop a three-marker microbial-related metabolite panel to assess 5-year risk of PaCa. The addition of five non-microbial metabolites further improves 5-year risk prediction of PaCa. The combined metabolite panel complements CA19-9, and individuals with a combined metabolite panel + CA19-9 score in the top 2.5th percentile have absolute 5-year risk estimates of >13%. The risk prediction model based on circulating microbial and non-microbial metabolites provides a potential tool to identify individuals at high risk of PaCa that would benefit from surveillance and/or from potential cancer interception strategies.
Collapse
Affiliation(s)
- Ehsan Irajizad
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ana Kenney
- Department of Statistics, University of California, Berkeley, Berkeley, CA, USA
| | - Tiffany Tang
- Department of Statistics, University of California, Berkeley, Berkeley, CA, USA
| | - Jody Vykoukal
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ranran Wu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eunice Murage
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer B Dennison
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marta Sans
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - James P Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maureen Loftus
- Dana-Farber Brigham and Women's Cancer Center, Division of Gastrointestinal Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - John A Chabot
- Division of Digestive and Liver Diseases, Columbia University Irving Medical Cancer and the Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Michael D Kluger
- Division of Digestive and Liver Diseases, Columbia University Irving Medical Cancer and the Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Fay Kastrinos
- Division of Digestive and Liver Diseases, Columbia University Irving Medical Cancer and the Vagelos College of Physicians and Surgeons, New York, NY, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Lauren Brais
- Dana-Farber Brigham and Women's Cancer Center, Division of Gastrointestinal Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ana Babic
- Dana-Farber Brigham and Women's Cancer Center, Division of Gastrointestinal Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kunal Jajoo
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Linda S Lee
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas E Clancy
- Dana-Farber Brigham and Women's Cancer Center, Division of Surgical Oncology, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA USA
| | - Kimmie Ng
- Dana-Farber Brigham and Women's Cancer Center, Division of Gastrointestinal Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Andrea Bullock
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jeanine M Genkinger
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA; Department of Epidemiology, Columbia Mailman School of Public Health, New York, NY, USA
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bin Yu
- Department of Statistics, University of California, Berkeley, Berkeley, CA, USA
| | - Brian M Wolpin
- Dana-Farber Brigham and Women's Cancer Center, Division of Gastrointestinal Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Sam Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Johannes F Fahrmann
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
45
|
Amri F, Belkhayat C, Yeznasni A, Koulali H, Jabi R, Zazour A, Abda N, Bouziane M, Ismaili Z, Kharrasse G. Association between pancreatic cancer and diabetes: insights from a retrospective cohort study. BMC Cancer 2023; 23:856. [PMID: 37697301 PMCID: PMC10496157 DOI: 10.1186/s12885-023-11344-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/28/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Studies investigating the prevalence of pancreatic cancer have revealed a heightened risk of 1.5 to 2.0 times among individuals with long-standing type 2 diabetes mellitus. AIMS We aimed to estimate the prevalence of diabetes among patients with pancreatic cancer, and identify the factors associated with type 2 diabetes mellitus in this population. METHODS This retrospective observational and analytical study was carried out in the Department of Gastroenterology of the Mohammed VI University Hospital over a period of 5 years, between 2018 and 2022, including all patients with confirmed cases of pancreatic adenocarcinoma. RESULTS Out of the 197 patients, 38.1% had a history of diabetes, among them, 42.7% had new-onset diabetes, while the remaining 57.3% had long-standing diabetes. Diabetic patients were significantly older than nondiabetic patients (mean age of 67.2 vs. 63, P = 0.009). Diabetes was more prevalent among obese patients (66.7%, P = 0.01), and less frequent among individuals with chronic alcohol consumption (20% vs. 80%, P = 0.04), and tobacco smokers (24.4% vs75.6%, P = 0.03). Among patients with an ECOG score ≥ 3, DM, 54.5% were DM-patients (P = 0.033). The same significant association was found for the Nutritional Risk Index, Patients who had moderate or severe malnutrition were more likely to be diabetic 74.7% (P = 0.004). Diabetic patients were less likely to undergo surgery due to comorbidities and general health deterioration. However, no significant differences were observed in sex, tumor stage or location. CONCLUSION Our study has shown an increased prevalence of diabetes in pancreatic cancer and highlights the importance of considering this cancer in cases of recent onset or uncontrolled diabetes, especially in elderly individuals.
Collapse
Affiliation(s)
- Fakhrddine Amri
- Department of Hepato-Gastroenterology, Mohammed VI University Hospital, Oujda University, Oujda, BP 4806, 60049, Morocco.
- Digestive Diseases Research Laboratory (DSRL), Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, Morocco.
| | - Chifaa Belkhayat
- Department of Hepato-Gastroenterology, Mohammed VI University Hospital, Oujda University, Oujda, BP 4806, 60049, Morocco
- Digestive Diseases Research Laboratory (DSRL), Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, Morocco
| | - Asmae Yeznasni
- Laboratory of Epidemiology, Clinical Research, and Public Health, Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, Morocco
| | - Hajar Koulali
- Department of Hepato-Gastroenterology, Mohammed VI University Hospital, Oujda University, Oujda, BP 4806, 60049, Morocco
- Digestive Diseases Research Laboratory (DSRL), Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, Morocco
| | - Rachid Jabi
- Department of General Surgery, Mohammed VI University Hospital, Oujda, Morocco
| | - Abdelkrim Zazour
- Department of Hepato-Gastroenterology, Mohammed VI University Hospital, Oujda University, Oujda, BP 4806, 60049, Morocco
- Digestive Diseases Research Laboratory (DSRL), Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, Morocco
| | - Naima Abda
- Laboratory of Epidemiology, Clinical Research, and Public Health, Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, Morocco
| | - Mohammed Bouziane
- Department of General Surgery, Mohammed VI University Hospital, Oujda, Morocco
| | - Zahi Ismaili
- Department of Hepato-Gastroenterology, Mohammed VI University Hospital, Oujda University, Oujda, BP 4806, 60049, Morocco
- Digestive Diseases Research Laboratory (DSRL), Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, Morocco
| | - Ghizlane Kharrasse
- Department of Hepato-Gastroenterology, Mohammed VI University Hospital, Oujda University, Oujda, BP 4806, 60049, Morocco
- Digestive Diseases Research Laboratory (DSRL), Faculty of Medicine and Pharmacy, Mohammed First University, Oujda, Morocco
| |
Collapse
|
46
|
Khalaf N, Kramer J, Liu Y, Abrams D, Singh H, El-Serag H, Kanwal F. Diabetes Status and Pancreatic Cancer Survival in the Nationwide Veterans Affairs Healthcare System. Dig Dis Sci 2023; 68:3634-3643. [PMID: 37474717 DOI: 10.1007/s10620-023-08035-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/03/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Long-standing type 2 diabetes is a known risk factor for developing pancreatic cancer, however, its influence on cancer-associated outcomes is understudied. AIMS To examine the associations between diabetes status and pancreatic cancer outcomes. METHODS We identified patients diagnosed with pancreatic adenocarcinoma in the national Veterans Administration System from 2010 to 2018. We classified each patient by pre-cancer diagnosis diabetes status: no diabetes, new-onset diabetes (NOD) of ≤ 3 years duration, or long-standing diabetes of > 3 years duration. We used Cox proportional hazards models to examine the association between diabetes status and survival. We adjusted the models for age, race, sex, body mass index, tobacco, and alcohol use, coronary artery disease, hypertension, chronic kidney disease, year of cancer diagnosis, and cancer stage and treatment. RESULTS We identified 6342 patients diagnosed with pancreatic adenocarcinoma. Most had long-standing diabetes (45.7%) prior to their cancer diagnosis, 14.5% had NOD, and 39.8% had no diabetes. Patients with long-standing diabetes had 10% higher mortality risk compared to patients without diabetes after adjusting for sociodemographic factors and medical comorbidities (adjusted HR 1.10; 95% CI 1.03-1.16). This difference in mortality remained statistically significant after additionally adjusting for cancer stage and receipt of potentially curative treatment (adjusted HR 1.09; 95% CI 1.02-1.15). There was no significant difference in mortality between patients with NOD compared to those without diabetes. CONCLUSIONS Long-standing but not new-onset diabetes is independently associated with increased mortality among patients with pancreatic cancer. This information has implication for prognostication and risk stratification among pancreatic cancer patients.
Collapse
Affiliation(s)
- Natalia Khalaf
- Center for Innovations in Quality, Effectiveness, and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Texas Medical Center Digestive Diseases Center, Houston, TX, USA.
- Michael E. DeBakey Veterans Affairs Medical Center, 2002 Holcombe Blvd. MS:111-D, Houston, TX, 77030, USA.
| | - Jennifer Kramer
- Center for Innovations in Quality, Effectiveness, and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
- Texas Medical Center Digestive Diseases Center, Houston, TX, USA
- Section of Health Services Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Yan Liu
- Center for Innovations in Quality, Effectiveness, and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Medical Center Digestive Diseases Center, Houston, TX, USA
| | - Daniela Abrams
- Department of Gastroenterology and Hepatology, University of Texas Medical Branch, Galveston, TX, USA
| | - Hardeep Singh
- Center for Innovations in Quality, Effectiveness, and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
- Michael E. DeBakey Veterans Affairs Medical Center, 2002 Holcombe Blvd. MS:111-D, Houston, TX, 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Hashem El-Serag
- Center for Innovations in Quality, Effectiveness, and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Medical Center Digestive Diseases Center, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Fasiha Kanwal
- Center for Innovations in Quality, Effectiveness, and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Medical Center Digestive Diseases Center, Houston, TX, USA
- Michael E. DeBakey Veterans Affairs Medical Center, 2002 Holcombe Blvd. MS:111-D, Houston, TX, 77030, USA
| |
Collapse
|
47
|
Jensen MH, Cichosz SL, Hejlesen O, Henriksen SD, Drewes AM, Olesen SS. Risk of pancreatic cancer in people with new-onset diabetes: A Danish nationwide population-based cohort study. Pancreatology 2023; 23:642-649. [PMID: 37422338 DOI: 10.1016/j.pan.2023.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/29/2023] [Accepted: 07/01/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND New onset diabetes (NOD) in people 50 years or older may indicate underlying pancreatic ductal adenocarcinoma (PDAC). The cumulative incidence of PDAC among people with NOD remains uncertain on a population-based level. METHODS This was a nationwide population-based retrospective cohort study based on the Danish national health registries. We investigated the 3-year cumulative incidence of PDAC in people 50 years or older with NOD. We further characterised people with pancreatic cancer-related diabetes (PCRD) in relation to demographic and clinical characteristics, including trajectories of routine biochemical parameters, using people with type 2 diabetes (T2D) as a comparator group. RESULTS During a 21-year observation period, we identified 353,970 people with NOD. Among them, 2105 people were subsequently diagnosed with pancreatic cancer within 3 years (0.59%, 95% CI [0.57-0.62%]). People with PCRD were older than people with T2D at diabetes diagnosis (median age 70.9 vs. 66.0 years (P < 0.001) and had a higher burden of comorbidities (P = 0.007) and more prescriptions of medications used to treat cardiovascular diseases (all P < 0.001). Distinct trajectories of HbA1c and plasma triglycerides were observed in PCRD vs. T2D, with group differences observed for up to three years prior to NOD diagnosis for HbA1c and up to two years for plasma triglyceride levels. CONCLUSIONS The 3-year cumulative incidence of PDAC is approximately 0.6% among people 50 years or older with NOD in a nationwide population-based setting. Compared to T2D, people with PCRD are characterised by distinct demographic and clinical profiles, including distinctive trajectories of plasma HbA1c and triglyceride levels.
Collapse
Affiliation(s)
- Morten Hasselstrøm Jensen
- Steno Diabetes Center North Denmark, Aalborg University Hospital, Aalborg, Denmark; Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Simon Lebech Cichosz
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Ole Hejlesen
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Stine Dam Henriksen
- Department of Gastrointestinal Surgery and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University Hospital, Aalborg, Denmark; Centre for Pancreatic Diseases and Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Asbjørn Mohr Drewes
- Steno Diabetes Center North Denmark, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University Hospital, Aalborg, Denmark; Centre for Pancreatic Diseases and Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Søren Schou Olesen
- Department of Clinical Medicine, Aalborg University Hospital, Aalborg, Denmark; Centre for Pancreatic Diseases and Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark.
| |
Collapse
|
48
|
Ohta H, Horii T, Yasu T. Adverse Metabolic Effects on Glucose in Patients Receiving Anamorelin Using a Japanese Claims Database. Oncology 2023; 101:782-785. [PMID: 37579746 DOI: 10.1159/000533539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 08/05/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND Anamorelin is the first drug approved for the treatment of cancer cachexia, a debilitating condition characterized by weight loss, anorexia, and muscle mass depletion. Cachexia negatively affects a patient's quality of life, survival, and response to chemotherapy. Studies describing anamorelin use are currently limited to a small number of pancreatic cancer cases. OBJECTIVES We aimed to examine the incidence and risk factors of adverse metabolic effects on glucose levels in cachexia patients with various carcinomas treated with anamorelin. METHOD We used real-world data of patients who received anamorelin between August 2021 and July 2022 and were registered in the JMDC claims database. We investigated the impact of metabolic adverse effects on glucose in patients receiving anamorelin with respect to the following factors: sex (male), age (>75 years), types of carcinoma, history of diabetes mellitus (DM), and concomitant use of steroids. RESULTS The incidence of adverse metabolic effects on glucose was 12.3%, and pancreatic cancer and history of DM were associated with adverse metabolic effects on glucose. The median onset of adverse metabolic effects on glucose was 17 days after anamorelin treatment. CONCLUSIONS This study highlights the need to monitor and manage hyperglycemia in cachexia patients receiving anamorelin, especially in those with pancreatic cancer and a history of DM.
Collapse
Affiliation(s)
- Hiroaki Ohta
- Department of Medicinal Therapy Research, Pharmaceutical Education and Research Center, Meiji Pharmaceutical University, Kiyose, Japan
| | - Takeshi Horii
- Department of Pharmacy, Faculty of Pharmacy, Musashino University, Nishitokyo, Japan
| | - Takeo Yasu
- Department of Medicinal Therapy Research, Pharmaceutical Education and Research Center, Meiji Pharmaceutical University, Kiyose, Japan
| |
Collapse
|
49
|
McDonnell D, Cheang AWE, Wilding S, Wild SH, Frampton AE, Byrne CD, Hamady ZZ. Elevated Glycated Haemoglobin (HbA1c) Is Associated with an Increased Risk of Pancreatic Ductal Adenocarcinoma: A UK Biobank Cohort Study. Cancers (Basel) 2023; 15:4078. [PMID: 37627106 PMCID: PMC10452109 DOI: 10.3390/cancers15164078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND The role of dysglycaemia as a risk marker for Pancreatic Ductal Adenocarcinoma (PDAC) is uncertain. We investigated the relationship between glycated haemoglobin (HbA1c) and incident PDAC using a retrospective cohort study within the UK Biobank. METHODS A study involving 499,804 participants from the UK Biobank study was undertaken. Participants were stratified by diabetes mellitus (DM) status, and then by HbA1c values < 42 mmol/mol, 42-47 mmol/mol, or ≥48 mmol/mol. Cox proportional hazard models were used to describe the association between HbA1c category (with time-varying interactions) and incident PDAC. RESULTS PDAC occurred in 1157 participants during 11.6 (10.9-12.3) years follow up [(median (interquartile range)]. In subjects without known DM at baseline, 12 months after recruitment, the adjusted hazard ratios (aHR, 95% CI) for incident PDAC for HbA1c 42-47 mmol/mol compared to HbA1c < 42 mmol/mol (reference group) was 2.10 (1.31-3.37, p = 0.002); and was 8.55 (4.58-15.99, p < 0.001) for HbA1c ≥ 48 mmol/mol. The association between baseline HbA1c and incident PDAC attenuated with increasing duration of time of follow-up to PDAC diagnosis. CONCLUSIONS Dysglycaemia detected by elevated HbA1c is associated with an increased risk of PDAC. The strength of the association between elevated HbA1c and incident PDAC is inversely proportional to the time from detecting dysglycaemia but remains significant for at least 60 months following HbA1c testing.
Collapse
Affiliation(s)
- Declan McDonnell
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
- HPB Unit, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Adrian W. E. Cheang
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
| | - Sam Wilding
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
| | - Sarah H. Wild
- Usher Institute, University of Edinburgh, Edinburgh EH8 9YL, UK;
| | - Adam E. Frampton
- Section of Oncology, University of Surrey, Guildford GU2 7XH, UK;
- HPB Unit, Royal Surrey NHS Foundation Trust, Guildford GU2 7XX, UK
| | - Christopher D. Byrne
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
| | - Zaed Z. Hamady
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
- HPB Unit, University Hospital Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
50
|
Bures J, Kohoutova D, Skrha J, Bunganic B, Ngo O, Suchanek S, Skrha P, Zavoral M. Diabetes Mellitus in Pancreatic Cancer: A Distinct Approach to Older Subjects with New-Onset Diabetes Mellitus. Cancers (Basel) 2023; 15:3669. [PMID: 37509329 PMCID: PMC10377806 DOI: 10.3390/cancers15143669] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/02/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is associated with a very poor prognosis, with near-identical incidence and mortality. According to the World Health Organization Globocan Database, the estimated number of new cases worldwide will rise by 70% between 2020 and 2040. There are no effective screening methods available so far, even for high-risk individuals. The prognosis of PDAC, even at its early stages, is still mostly unsatisfactory. Impaired glucose metabolism is present in about 3/4 of PDAC cases. METHODS Available literature on pancreatic cancer and diabetes mellitus was reviewed using a PubMed database. Data from a national oncology registry (on PDAC) and information from a registry of healthcare providers (on diabetes mellitus and a number of abdominal ultrasound investigations) were obtained. RESULTS New-onset diabetes mellitus in subjects older than 60 years should be an incentive for a prompt and detailed investigation to exclude PDAC. Type 2 diabetes mellitus, diabetes mellitus associated with chronic non-malignant diseases of the exocrine pancreas, and PDAC-associated type 3c diabetes mellitus are the most frequent types. Proper differentiation of particular types of new-onset diabetes mellitus is a starting point for a population-based program. An algorithm for subsequent steps of the workup was proposed. CONCLUSIONS The structured, well-differentiated, and elaborately designed approach to the elderly with a new onset of diabetes mellitus could improve the current situation in diagnostics and subsequent poor outcomes of therapy of PDAC.
Collapse
Affiliation(s)
- Jan Bures
- Institute of Gastrointestinal Oncology, Military University Hospital Prague, 169 02 Prague, Czech Republic
- Department of Medicine, First Faculty of Medicine, Charles University, Prague and Military University Hospital Prague, 169 02 Prague, Czech Republic
- Biomedical Research Centre, University Hospital Hradec Kralove, 500 03 Hradec Kralove, Czech Republic
| | - Darina Kohoutova
- Biomedical Research Centre, University Hospital Hradec Kralove, 500 03 Hradec Kralove, Czech Republic
- The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK
| | - Jan Skrha
- Third Department of Internal Medicine-Endocrinology and Metabolism, First Faculty of Medicine, Charles University, Prague and General University Hospital in Prague, 128 08 Prague, Czech Republic
| | - Bohus Bunganic
- Department of Medicine, First Faculty of Medicine, Charles University, Prague and Military University Hospital Prague, 169 02 Prague, Czech Republic
| | - Ondrej Ngo
- Institute of Health Information and Statistics of the Czech Republic, 128 01 Prague, Czech Republic
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, 602 00 Brno, Czech Republic
| | - Stepan Suchanek
- Institute of Gastrointestinal Oncology, Military University Hospital Prague, 169 02 Prague, Czech Republic
- Department of Medicine, First Faculty of Medicine, Charles University, Prague and Military University Hospital Prague, 169 02 Prague, Czech Republic
| | - Pavel Skrha
- Department of Medicine, Third Faculty of Medicine, Charles University, Prague and University Hospital Kralovske Vinohrady, 100 00 Prague, Czech Republic
| | - Miroslav Zavoral
- Institute of Gastrointestinal Oncology, Military University Hospital Prague, 169 02 Prague, Czech Republic
- Department of Medicine, First Faculty of Medicine, Charles University, Prague and Military University Hospital Prague, 169 02 Prague, Czech Republic
| |
Collapse
|