1
|
Farinella R, Felici A, Peduzzi G, Testoni SGG, Costello E, Aretini P, Blazquez-Encinas R, Oz E, Pastore A, Tacelli M, Otlu B, Campa D, Gentiluomo M. From classical approaches to artificial intelligence, old and new tools for PDAC risk stratification and prediction. Semin Cancer Biol 2025; 112:71-92. [PMID: 40147701 DOI: 10.1016/j.semcancer.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is recognized as one of the most lethal malignancies, characterized by late-stage diagnosis and limited therapeutic options. Risk stratification has traditionally been performed using epidemiological studies and genetic analyses, through which key risk factors, including smoking, diabetes, chronic pancreatitis, and inherited predispositions, have been identified. However, the multifactorial nature of PDAC has often been insufficiently addressed by these methods, leading to limited precision in individualized risk assessments. Advances in artificial intelligence (AI) have been proposed as a transformative approach, allowing the integration of diverse datasets-spanning genetic, clinical, lifestyle, and imaging data into dynamic models capable of uncovering novel interactions and risk profiles. In this review, the evolution of PDAC risk stratification is explored, with classical epidemiological frameworks compared to AI-driven methodologies. Genetic insights, including genome-wide association studies and polygenic risk scores, are discussed, alongside AI models such as machine learning, radiomics, and deep learning. Strengths and limitations of these approaches are evaluated, with challenges in clinical translation, such as data scarcity, model interpretability, and external validation, addressed. Finally, future directions are proposed for combining classical and AI-driven methodologies to develop scalable, personalized predictive tools for PDAC, with the goal of improving early detection and patient outcomes.
Collapse
Affiliation(s)
| | | | | | - Sabrina Gloria Giulia Testoni
- Division of Gastroenterology and Gastrointestinal Endoscopy, IRCCS Policlinico San Donato, Vita-Salute San Raffaele University, Milan, Italy
| | - Eithne Costello
- Liverpool Experimental Cancer Medicine Centre, University of Liverpool, Liverpool, United Kingdom
| | - Paolo Aretini
- Fondazione Pisana per la Scienza, San Giuliano Terme, Italy
| | - Ricardo Blazquez-Encinas
- Department of Cell Biology, Physiology and Immunology, University of Cordoba / Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Elif Oz
- Department of Biostatistics and Bioinformatics, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Aldo Pastore
- Fondazione Pisana per la Scienza, San Giuliano Terme, Italy
| | - Matteo Tacelli
- Pancreas Translational & Clinical Research Center, Pancreato-Biliary Endoscopy and Endosonography Division, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Burçak Otlu
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, Ankara, Turkey
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | | |
Collapse
|
2
|
Tan JT, Mao X, Cheng HM, Seto WK, Leung WK, Cheung KS. Aspirin is associated with lower risk of pancreatic cancer and cancer-related mortality in patients with diabetes mellitus. Gut 2025; 74:603-612. [PMID: 39746785 DOI: 10.1136/gutjnl-2024-333329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Patients with type 2 diabetes mellitus (T2DM) have higher pancreatic cancer (PC) risk. While aspirin has chemopreventive effects on digestive cancers, its effect on PC among patients with T2DM is unclear. METHODS This retrospective cohort study identified newly diagnosed adult patients with T2DM in Hong Kong between 2001 and 2015 from a territory-wide healthcare registry. Exclusion criteria were history of PC, pancreatic cyst, IgG4 disease, or pancreatectomy. To address reverse causality between PC and T2DM, we excluded patients with PC diagnosed within 1 year of T2DM. We also excluded patients with less than 1 year of observation. Primary outcome was PC, and secondary outcomes were PC-related and all-cause mortality. Aspirin use was treated as time-varying variable (≥180 day-use/year) to address immortal-time bias, and multivariable Cox regression model was employed to derive adjusted HR (aHR). Propensity-score (PS) matching was used as secondary analysis. RESULTS Among 343 966 newly diagnosed patients with T2DM (median follow-up: 10.5 years; IQR: 7.7-14.5 years), 1224 (0.36%) developed PC. There were 51 151 (14.9%) deaths from any cause, and 787 (0.2%) died from PC. Aspirin use was associated with lower PC risk in both time-dependent (aHR: 0.58; 95% CI 0.49 to 0.69) and PS matching analysis (aHR: 0.61; 95% CI 0.48 to 0.77). An inverse relationship was observed with increasing dose and duration of aspirin use (P trend<0.001). Aspirin was also associated with a lower risk of PC-related mortality (aHR: 0.43; 95% CI 0.34 to 0.53) and all-cause mortality (aHR: 0.78; 95% CI 0.76 to 0.80). CONCLUSION Aspirin use may be an oncopreventive strategy to reduce PC risk in patients with T2DM. Further studies are warranted to validate the study findings.
Collapse
Affiliation(s)
- Jing Tong Tan
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Xianhua Mao
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ho-Ming Cheng
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Wai-Kay Seto
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Wai-K Leung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Ka-Shing Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
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 PMCID: PMC11960203 DOI: 10.4093/dmj.2024.0277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
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
|
5
|
Ying H, Kimmelman AC, Bardeesy N, Kalluri R, Maitra A, DePinho RA. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev 2025; 39:36-63. [PMID: 39510840 PMCID: PMC11789498 DOI: 10.1101/gad.351863.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a grim prognosis for patients. Recent multidisciplinary research efforts have provided critical insights into its genetics and tumor biology, creating the foundation for rational development of targeted and immune therapies. Here, we review the PDAC genomic landscape and the role of specific oncogenic events in tumor initiation and progression, as well as their contributions to shaping its tumor biology. We further summarize and synthesize breakthroughs in single-cell and metabolic profiling technologies that have illuminated the complex cellular composition and heterotypic interactions of the PDAC tumor microenvironment, with an emphasis on metabolic cross-talk across cancer and stromal cells that sustains anabolic growth and suppresses tumor immunity. These conceptual advances have generated novel immunotherapy regimens, particularly cancer vaccines, which are now in clinical testing. We also highlight the advent of KRAS targeted therapy, a milestone advance that has transformed treatment paradigms and offers a platform for combined immunotherapy and targeted strategies. This review provides a perspective summarizing current scientific and therapeutic challenges as well as practice-changing opportunities for the PDAC field at this major inflection point.
Collapse
Affiliation(s)
- Haoqiang Ying
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA;
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Alec C Kimmelman
- Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, New York 10016, USA
- Department of Radiation Oncology, New York University Grossman School of Medicine, New York, New York 10016, USA
| | - Nabeel Bardeesy
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts 02114, USA
- The Cancer Program, Broad Institute, Cambridge, Massachusetts 02142, USA
| | - Raghu Kalluri
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Bioengineering, Rice University, Houston, Texas 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Anirban Maitra
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
- Sheikh Ahmed Pancreatic Cancer Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ronald A DePinho
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas 77030, USA;
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
6
|
Egan AM, Wood-Wentz CM, Mohan S, Bailey KR, Vella A. Baseline Fasting Glucose Level, Age, Sex, and Body Mass Index and the Development of Diabetes in US Adults. JAMA Netw Open 2025; 8:e2456067. [PMID: 39847352 PMCID: PMC11758592 DOI: 10.1001/jamanetworkopen.2024.56067] [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: 08/30/2024] [Accepted: 11/18/2024] [Indexed: 01/24/2025] Open
Abstract
Importance Understanding the interplay between diabetes risk factors and diabetes development is important to develop individual, practice, and population-level prevention strategies. Objective To evaluate the progression from normal and impaired fasting glucose levels to diabetes among adults. Design, Setting, and Participants This retrospective community-based cohort study used data from the Rochester Epidemiology Project, in Olmsted County, Minnesota, on 44 992 individuals with at least 2 fasting plasma glucose (FPG) measurements from January 1, 2005, to December 31, 2017. People who met criteria for diabetes on or before their first FPG measurement were excluded. Data were electronically retrieved in December 2019 with analyses finalized in November 2024. Exposures The exposure was baseline FPG level, with covariates including the following measures that are consistently recorded in the electronic health record: body mass index (BMI), age, and sex. Main Outcomes and Measures The cumulative probability of freedom from diabetes was estimated and presented graphically using a Kaplan-Meier curve. Multivariable Cox proportional hazards regression modeling was used to estimate the partial hazard ratios (HRs) for variables of interest. Diabetes was defined as an FPG level greater than 125 mg/dL. Results A total of 44 992 individuals (mean [SD] age at baseline, 43.7 [11.8] years; 26 025 women [57.8%]) were included. The baseline mean (SD) BMI was 28.9 (6.6). Over a median follow-up of 6.8 years (IQR, 3.6-9.7 years), 3879 individuals (8.6%) developed diabetes. The Kaplan-Meier 10-year cumulative risk of incident diabetes was 12.8% (95% CI, 12.4%-13.2%). All initial FPG levels outside a range of 80 to 94 mg/dL were associated with increased risk for diabetes (ie, FPG <70 mg/dL: HR, 3.49 [95% CI, 2.19-5.57]; FPG 120-125 mg/dL: HR, 12.47 [10.84-14.34]). Other independent risk factors were male sex (HR, 1.31 [95% CI, 1.22-1.40]), older age (≥60 years: HR, 1.97 [95% CI, 1.71-2.28]), and any abnormal category of BMI, including underweight (BMI <18.5: HR, 2.42 [95% CI, 1.77-3.29]; BMI ≥40: HR, 4.03 [95% CI, 3.56-4.56]). There was a significant additive association of variables, particularly FPG level and BMI. For instance, a woman aged 55 to 59 years with a BMI of 18.5 to 24.9 and an FPG level of 95 to 99 mg/dL had an estimated 10-year diabetes risk of 7.0%. However, an almost doubling of risk to 13.0% was observed if the BMI was 30.0 to 34.9, and risk more than doubled again to 28.0% if FPG level also increased to 105 to 109 mg/dL. A nomogram was generated to facilitate individual classification into one of four 10-year risk categories. Conclusions and Relevance This retrospective cohort study of 44 992 individuals suggests that FPG level, age, BMI, and male sex were all associated with development of diabetes, with significant interaction between these variables. These data contribute to understanding the clinical course of diabetes and highlight the substantial individual variation in diabetes risk according to commonly measured clinical variables. The findings facilitate lifestyle and pharmacologic interventions to treat those at highest risk of diabetes to reduce future morbidity and mortality. Further work is needed to validate this risk categorization tool for different populations.
Collapse
Affiliation(s)
- Aoife M. Egan
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, Minnesota
| | | | - Sneha Mohan
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, Minnesota
| | - Kent R. Bailey
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, Minnesota
| | - Adrian Vella
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
7
|
Katona BW, Lubinski J, Pal T, Huzarski T, Foulkes WD, Moller P, Eisen A, Randall Armel S, Neuhausen SL, Raj R, Aeilts A, Singer CF, Bordeleau L, Karlan B, Olopade O, Tung N, Zakalik D, Kotsopoulos J, Fruscio R, Eng C, Sun P, Narod SA. The incidence of pancreatic cancer in women with a BRCA1 or BRCA2 mutation. Cancer 2025; 131:e35666. [PMID: 39611336 DOI: 10.1002/cncr.35666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/17/2024] [Accepted: 08/12/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND The lifetime risk of pancreatic cancer in women with a germline mutation in BRCA1 and BRCA2 is not well established. In an international prospective cohort of female carriers of BRCA1 and BRCA2 mutations, the cumulative incidence of pancreatic cancer from age 40 until 80 years was estimated. METHODS A total of 8295 women with a BRCA1 or BRCA2 mutation were followed for new cases of pancreatic cancer. Subjects were followed from the date of baseline questionnaire or age 40 years (whichever came last) until a new diagnosis of pancreatic cancer, death from another cause, or date of last follow-up. RESULTS Thirty-four incident pancreatic cancer cases were identified in the cohort. The annual risk of pancreatic cancer between age 40 and 80 years was 0.04% for BRCA1 carriers and 0.09% for BRCA2 carriers. Via the Kaplan-Meier method, the cumulative incidence from age 40 to 80 years was 2.2% (95% CI, 1.1%-4.3%) for BRCA1 carriers and 2.7% (95% CI, 1.3%-5.4%) for BRCA2 carriers. Only two of the 34 cases reported a first-degree relative with pancreatic cancer (hazard ratio, 4.75; 95% CI, 1.13-19.9; p = .03). Risk factors for pancreatic cancer included alcohol intake and a history of diabetes. The 5-year survival rate for the 34 cases was 8.8%. CONCLUSIONS The lifetime risk of pancreatic cancer is approximately 2% in women with a BRCA1 mutation and 3% for women with a BRCA2 mutation. The poor survival in hereditary pancreatic cancer underscores the need for novel antitumoral strategies.
Collapse
Affiliation(s)
- Bryson W Katona
- Basser Center for BRCA and Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jan Lubinski
- International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Tuya Pal
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tomasz Huzarski
- International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - William D Foulkes
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Pal Moller
- Institute of Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Andrea Eisen
- Sunnybrook Regional Cancer Centre, Toronto, Ontario, Canada
| | - Susan Randall Armel
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
- Division of Gynecologic Oncology, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Susan L Neuhausen
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - Rebecca Raj
- Women's College Research Institute, Toronto, Ontario, Canada
| | - Amber Aeilts
- Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, Ohio, USA
| | - Christian F Singer
- Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Louise Bordeleau
- Department of Oncology, Juravinski Cancer Centre, Hamilton, Ontario, Canada
| | - Beth Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Olufunmilayo Olopade
- Department of Medicine and Human Genetics, University of Chicago, Chicago, Illinois, USA
| | - Nadine Tung
- Cancer Risk and Prevention Program, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Dana Zakalik
- Cancer Genetics Program, Beaumont Hospital, Royal Oak, Michigan, USA
| | - Joanne Kotsopoulos
- Women's College Research Institute, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Robert Fruscio
- Department of Medicine and Surgery, University of Milan Bicocca, IRCCS San Gerardo, Monza, Italy
| | - Charis Eng
- Center for Personalized Genetic Healthcare, Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ping Sun
- Women's College Research Institute, Toronto, Ontario, Canada
| | - Steven A Narod
- Women's College Research Institute, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Gong J, Li X, Feng Z, Lou J, Pu K, Sun Y, Hu S, Zhou Y, Song T, Shangguan M, Zhang K, Lu W, Dong X, Wu J, Zhu H, He Q, Xu H, Wu Y. Sorcin can trigger pancreatic cancer-associated new-onset diabetes through the secretion of inflammatory cytokines such as serpin E1 and CCL5. Exp Mol Med 2024; 56:2535-2547. [PMID: 39516378 PMCID: PMC11612510 DOI: 10.1038/s12276-024-01346-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 07/28/2024] [Accepted: 08/19/2024] [Indexed: 11/16/2024] Open
Abstract
A rise in blood glucose is an early warning sign of underlying pancreatic cancer (PC) and may be an indicator of genetic events in PC progression. However, there is still a lack of mechanistic research on pancreatic cancer-associated new-onset diabetes (PCAND). In the present study, we identified a gene SRI, which possesses a SNP with the potential to distinguish PCAND and Type 2 diabetes mellitus (T2DM), by machine learning on the basis of the UK Biobank database. In vitro and in vivo, sorcin overexpression induced pancreatic β-cell dysfunction. Sorcin can form a positive feedback loop with STAT3 to increase the transcription of serpin E1 and CCL5, which may directly induce β-cell dysfunction. In 88 biopsies, the expression of sorcin was elevated in PC tissues, especially in PCAND samples. Furthermore, plasma serpin E1 levels are higher in peripheral blood samples from PCAND patients than in those from T2DM patients. In conclusion, sorcin may be the key driver in PCAND, and further study on the sorcin-STAT3-serpin E1/CCL5 signaling axis may help us better understand the pathogenesis of PCAND and identify potential biomarkers.
Collapse
Affiliation(s)
- Jiali Gong
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Xiawei Li
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Zengyu Feng
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianyao Lou
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kaiyue Pu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yongji Sun
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Sien Hu
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Yizhao Zhou
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tianyu Song
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Meihua Shangguan
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kai Zhang
- School of Public Health and Eye Center The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Wenjie Lu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xin Dong
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jian Wu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Wenzhou, Zhejiang University, Wenzhou, Zhejiang, China
| | - Hong Zhu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiaojun He
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
- Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, Zhejiang, China.
| | - Hongxia Xu
- Innovation Institute for Artificial Intelligence in Medicine and Liangzhu Laboratory, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Yulian Wu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
- Department of Surgery, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China.
| |
Collapse
|
11
|
Eze-Odurukwe A, Rehman A, Ayinla L, Anika NN, Shahid R, Ugwuoru AL, Mansoor M, Kamran M. Metabolite Biomarkers for Early Detection of Pancreatic Ductal Adenocarcinoma: A Systematic Review. Cureus 2024; 16:e74528. [PMID: 39726485 PMCID: PMC11671176 DOI: 10.7759/cureus.74528] [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] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with a poor prognosis. This poor prognosis is largely attributed to a late-stage diagnosis. Recent advancements in metabolomics have emerged as a promising avenue for biomarker discovery in PDAC. This systematic review evaluates the potential of metabolite biomarkers for early detection of PDAC. Four studies meeting the inclusion criteria were analyzed, encompassing experimental, case-control, and prospective cohort designs. Key findings include the identification of distinct metabolic subtypes in PDAC with varying sensitivities to metabolic inhibitors. A biomarker signature comprising nine metabolites plus CA19-9 showed high accuracy in distinguishing PDAC from chronic pancreatitis, outperforming CA19-9 alone. Another study identified a five-metabolite signature demonstrating high diagnostic accuracy for pancreatic cancer, differentiating it from type 2 diabetes mellitus. A two-metabolite model (isoleucine and adrenic acid) showed superior performance in detecting stage-I PDAC compared to CA19-9. These studies consistently demonstrate altered metabolic pathways in PDAC patients compared to healthy controls and those with benign pancreatic conditions. Integrating metabolomic data with other molecular profiling approaches has become a powerful strategy for improving diagnostic accuracy. However, challenges remain, including the influence of confounding factors, the need for large-scale validation studies, and the standardization of metabolomic methods. The potential of artificial intelligence in interpreting complex metabolomic data offers promising avenues for future research. This review highlights the significant potential of metabolite biomarkers in early PDAC detection while emphasizing the need for further validation and refinement of these approaches.
Collapse
Affiliation(s)
| | | | - Lois Ayinla
- General Medicine, All Saints University School of Medicine, Roseau, DMA
| | - Nabila N Anika
- Surgery, Baylor College of Medicine, Houston, USA
- Medicine and Surgery, Holy Family Red Crescent Medical College and Hospital, Dhaka, BGD
| | - Ramsha Shahid
- Physiology, Akhtar Saeed Medical and Dental College, Islamabad, PAK
| | | | - Muzafar Mansoor
- Internal Medicine, Allama Iqbal Medical College, Lahore, PAK
| | | |
Collapse
|
12
|
Gong J, Zhang Y, Guang Z, Rahaman A, Xue J, Wang Q, Yao Y, Wang P, Wang Y. Single hollow-core microstructure fiber biosensor for pancreatic cancer diagnosis in terahertz regime. PHYSICA SCRIPTA 2024; 99:115505. [DOI: 10.1088/1402-4896/ad7f00] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Abstract
Pancreatic cancer is a kind of malignant tumor that is difficult to detect in its early stages, developing rapidly and with a 5-year survival rate of only 5% to 10%. Therefore early diagnosis and discovery of pancreatic cancer are very important for the successful treatment of the disease. Here, we report a single hollow-core microstructural fiber (SHC-MSF) biosensor based on a ZEONEX substrate, which has been optimized for the early detection of pancreatic cancer biomarkers. The proposed SHC-MSF biosensor adopts a single-aperture structure to increase the contact range with assay analytes to improve the detection sensitivity. Its biosensing performance was numerically analyzed using a finite element method with a perfect matching layer. Numerical results demonstrated that the proposed MSF-biosensor presented ultra-high sensitivity (bilirubin: 105.55%, glucose: 105.34%, creatinine: 105.67%) and negligible confinement loss (bilirubin: 5.52 × 10−14 cm−1, glucose: 1.65 × 10−14 cm−1, creatinine: 5.57 × 10−14 cm−1) in the range of 0.3 ∼ 2.0 THz. Moreover, the SHC-MSF biosensor could selectively detect and distinguish cancer markers of different concentrations in the blood to achieve a more accurate diagnosis of pancreatic cancer. Finally, fabrication tolerance analysis of the proposed MSF-biosensor is provided to ensure the feasibility of rapid preparation.
Collapse
|
13
|
Borgmästars E, Ulfenborg B, Johansson M, Jonsson P, Billing O, Franklin O, Lundin C, Jacobson S, Simm M, Lubovac-Pilav Z, Sund M. Multi-omics profiling to identify early plasma biomarkers in pre-diagnostic pancreatic ductal adenocarcinoma: a nested case-control study. Transl Oncol 2024; 48:102059. [PMID: 39018772 PMCID: PMC11301391 DOI: 10.1016/j.tranon.2024.102059] [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: 04/05/2024] [Revised: 05/20/2024] [Accepted: 07/05/2024] [Indexed: 07/19/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with poor survival. Novel biomarkers are urgently needed to improve the outcome through early detection. Here, we aimed to discover novel biomarkers for early PDAC detection using multi-omics profiling in pre-diagnostic plasma samples biobanked after routine health examinations. A nested case-control study within the Northern Sweden Health and Disease Study was designed. Pre-diagnostic plasma samples from 37 future PDAC patients collected within 2.3 years before diagnosis and 37 matched healthy controls were included. We analyzed metabolites using liquid chromatography mass spectrometry and gas chromatography mass spectrometry, microRNAs by HTG edgeseq, proteins by multiplex proximity extension assays, as well as three clinical biomarkers using milliplex technology. Supervised and unsupervised multi-omics integration were performed as well as univariate analyses for the different omics types and clinical biomarkers. Multiple hypothesis testing was corrected using Benjamini-Hochberg's method and a false discovery rate (FDR) below 0.1 was considered statistically significant. Carbohydrate antigen (CA) 19-9 was associated with PDAC risk (OR [95 % CI] = 3.09 [1.31-7.29], FDR = 0.03) and increased closer to PDAC diagnosis. Supervised multi-omics models resulted in poor discrimination between future PDAC cases and healthy controls with obtained accuracies between 0.429-0.500. No single metabolite, microRNA, or protein was differentially altered (FDR < 0.1) between future PDAC cases and healthy controls. CA 19-9 levels increase up to two years prior to PDAC diagnosis but extensive multi-omics analysis including metabolomics, microRNAomics and proteomics in this cohort did not identify novel early biomarkers for PDAC.
Collapse
Affiliation(s)
- Emmy Borgmästars
- Department of Diagnostics and Intervention/ Surgery, Umeå University, Umeå, Sweden.
| | - Benjamin Ulfenborg
- School of Bioscience, Department of Biology and Bioinformatics, University of Skövde, Skövde, Sweden
| | - Mattias Johansson
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Pär Jonsson
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Ola Billing
- Department of Diagnostics and Intervention/ Surgery, Umeå University, Umeå, Sweden
| | - Oskar Franklin
- Department of Diagnostics and Intervention/ Surgery, Umeå University, Umeå, Sweden; Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Aurora, CO, USA
| | - Christina Lundin
- Department of Diagnostics and Intervention/ Surgery, Umeå University, Umeå, Sweden
| | - Sara Jacobson
- Department of Diagnostics and Intervention/ Surgery, Umeå University, Umeå, Sweden
| | - Maja Simm
- Department of Diagnostics and Intervention/ Surgery, Umeå University, Umeå, Sweden; Department of Clinical Sciences/ Obstetrics and Gynecology, Umeå University, Umeå, Sweden
| | - Zelmina Lubovac-Pilav
- School of Bioscience, Department of Biology and Bioinformatics, University of Skövde, Skövde, Sweden
| | - Malin Sund
- Department of Diagnostics and Intervention/ Surgery, Umeå University, Umeå, Sweden; Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
14
|
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
|
15
|
Pita-Grisanti V, Velez-Bonet E, Chasser K, Hurst Z, Liette A, Vulic G, Dubay K, Lahooti A, Badi N, Ueltschi O, Gumpper-Fedus K, Hsueh HY, Lahooti I, Chavez-Tomar M, Terhorst S, Knoblaugh SE, Cao L, Huang W, Coss CC, Mace TA, Choueiry F, Hinton A, Culp S, Mitchell JM, Schmandt R, Grinsfelder MO, Basen-Engquist K, Cruz-Monserrate Z. Physical Activity Decreases Inflammation and Delays the Development of Obesity-Associated Pancreatic Ductal Adenocarcinoma. Cancer Res 2024; 84:3058-3071. [PMID: 38781455 PMCID: PMC11405134 DOI: 10.1158/0008-5472.can-23-1045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 02/29/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
Obesity is a risk factor for pancreatic ductal adenocarcinoma (PDAC), a deadly disease with limited preventive strategies. Lifestyle interventions to decrease obesity represent a potential approach to prevent obesity-associated PDAC. In this study, we examined whether decreasing obesity through physical activity (PA) and/or dietary changes could decrease inflammation in humans and prevent obesity-associated PDAC in mice. Comparison of circulating inflammatory-associated cytokines in subjects (overweight and obese) before and after a PA intervention revealed PA lowered systemic inflammatory cytokines. Mice with pancreatic-specific inducible KrasG12D expression were exposed to PA and/or dietary interventions during and after obesity-associated cancer initiation. In mice with concurrent diet-induced obesity and KrasG12D expression, the PA intervention led to lower weight gain, suppressed systemic inflammation, delayed tumor progression, and decreased proinflammatory signals in the adipose tissue. However, these benefits were not as evident when obesity preceded pancreatic KrasG12D expression. Combining PA with diet-induced weight loss (DI-WL) delayed obesity-associated PDAC progression in the genetically engineered mouse model, but neither PA alone nor combined with DI-WL or chemotherapy prevented PDAC tumor growth in orthotopic PDAC models regardless of obesity status. PA led to the upregulation of Il15ra in adipose tissue. Adipose-specific overexpression of Il15 slowed PDAC growth but only in nonobese mice. Overall, our study suggests that PA alone or combined with DI-WL can reduce inflammation and delay obesity-associated PDAC development or progression. Lifestyle interventions that prevent or manage obesity or therapies that target weight loss-related molecular pathways could prevent progression of PDAC. Significance: Physical activity reduces inflammation and induces changes to adipose-related signaling to suppress pancreatic cancer, supporting the potential of obesity management strategies to reduce the risk of developing pancreatic cancer. See related commentary by Sogunro and Muzumdar, p. 2935.
Collapse
Affiliation(s)
- Valentina Pita-Grisanti
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- The Ohio State University Interdisciplinary Nutrition Program, The Ohio State University, Columbus, OH
| | - Ericka Velez-Bonet
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- The Ohio State University Interdisciplinary Nutrition Program, The Ohio State University, Columbus, OH
| | - Kaylin Chasser
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Zachary Hurst
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- The Ohio State University Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH
| | - Alexus Liette
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Grace Vulic
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Kelly Dubay
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Ali Lahooti
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Niharika Badi
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Olivia Ueltschi
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Kristyn Gumpper-Fedus
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Hsiang-Yin Hsueh
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- The Ohio State University Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH
| | - Ila Lahooti
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Myrriah Chavez-Tomar
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Samantha Terhorst
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Sue E. Knoblaugh
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH
| | - Lei Cao
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Wei Huang
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Christopher C. Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Thomas A. Mace
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Fouad Choueiry
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Alice Hinton
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, OH
| | - Stacey Culp
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Jennifer M Mitchell
- Department of Veterinary Medicine and Surgery, UT MD Anderson Cancer Center, Houston, TX
| | - Rosemarie Schmandt
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, UT MD Anderson Cancer Center, Houston, TX
| | - Michaela Onstad Grinsfelder
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, UT MD Anderson Cancer Center, Houston, TX
| | - Karen Basen-Engquist
- Department of Behavioral Science, Center for Energy Balance, The University of Texas MD Anderson Cancer Center, UT MD Anderson Cancer Center, Houston, TX
| | - Zobeida Cruz-Monserrate
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| |
Collapse
|
16
|
Klatte DCF, Weston A, Ma Y, Sledge H, Bali A, Bolan C, Engels M, van Hooft JE, van Leerdam ME, Ouni A, Wallace MB, Bi Y. Temporal Trends in Body Composition and Metabolic Markers Prior to Diagnosis of Pancreatic Ductal Adenocarcinoma. Clin Gastroenterol Hepatol 2024; 22:1830-1838.e9. [PMID: 38703880 DOI: 10.1016/j.cgh.2024.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND & AIMS Changes in body composition and metabolic factors may serve as biomarkers for the early detection of pancreatic ductal adenocarcinoma (PDAC). The aim of this study was to capture the longitudinal changes in body composition and metabolic factors before diagnosis of PDAC. METHODS We performed a retrospective cohort study in which all patients (≥18 years) diagnosed with PDAC from 2002 to 2021 were identified. We collected all abdominal computed tomography scans and 10 different blood-based biomarkers up to 36 months before diagnosis. We applied a fully automated abdominal segmentation algorithm previously developed by our group for 3-dimensional quantification of body composition on computed tomography scans. Longitudinal trends of body composition and blood-based biomarkers before PDAC diagnosis were estimated using linear mixed models, compared across different time windows, and visualized using spline regression. RESULTS We included 1690 patients in body composition analysis, of whom 516 (30.5%) had ≥2 prediagnostic computed tomography scans. For analysis of longitudinal trends of blood-based biomarkers, 3332 individuals were included. As an early manifestation of PDAC, we observed a significant decrease in visceral and subcutaneous adipose tissue (β = -1.94 [95% confidence interval (CI), -2.39 to -1.48] and β = -2.59 [95% CI, -3.17 to -2.02]) in area (cm2)/height (m2) per 6 months closer to diagnosis, accompanied by a decrease in serum lipids (eg, low-density lipoprotein [β = -2.83; 95% CI, -3.31 to -2.34], total cholesterol [β = -2.69; 95% CI, -3.18 to -2.20], and triglycerides [β = -1.86; 95% CI, -2.61 to -1.11]), and an increase in blood glucose levels. Loss of muscle tissue and bone volume was predominantly observed in the last 6 months before diagnosis. CONCLUSIONS This study identified significant alterations in a variety of soft tissue and metabolic markers that occur in the development of PDAC. Early recognition of these metabolic changes may provide an opportunity for early detection.
Collapse
Affiliation(s)
- Derk C F Klatte
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Alexander Weston
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Yaohua Ma
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Hanna Sledge
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Aman Bali
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | - Candice Bolan
- Department of Radiology, Mayo Clinic, Jacksonville, Florida
| | - Megan Engels
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeanin E van Hooft
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands; Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ahmed Ouni
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | - Michael B Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | - Yan Bi
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
17
|
Cheng HH, Shevach JW, Castro E, Couch FJ, Domchek SM, Eeles RA, Giri VN, Hall MJ, King MC, Lin DW, Loeb S, Morgan TM, Offit K, Pritchard CC, Schaeffer EM, Szymaniak BM, Vassy JL, Katona BW, Maxwell KN. BRCA1, BRCA2, and Associated Cancer Risks and Management for Male Patients: A Review. JAMA Oncol 2024; 10:1272-1281. [PMID: 39052257 DOI: 10.1001/jamaoncol.2024.2185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Importance Half of all carriers of inherited cancer-predisposing variants in BRCA1 and BRCA2 are male, but the implications for their health are underrecognized compared to female individuals. Germline variants in BRCA1 and BRCA2 (also known as pathogenic or likely pathogenic variants, referred to here as BRCA1/2 PVs) are well known to significantly increase the risk of breast and ovarian cancers in female carriers, and knowledge of BRCA1/2 PVs informs established cancer screening and options for risk reduction. While risks to male carriers of BRCA1/2 PVs are less characterized, there is convincing evidence of increased risk for prostate cancer, pancreatic cancer, and breast cancer in males. There has also been a rapid expansion of US Food and Drug Administration-approved targeted cancer therapies, including poly ADP ribose polymerase (PARP) inhibitors, for breast, pancreatic, and prostate cancers associated with BRCA1/2 PVs. Observations This narrative review summarized the data that inform cancer risks, targeted cancer therapy options, and guidelines for early cancer detection. It also highlighted areas of emerging research and clinical trial opportunities for male BRCA1/2 PV carriers. These developments, along with the continued relevance to family cancer risk and reproductive options, have informed changes to guideline recommendations for genetic testing and strengthened the case for increased genetic testing for males. Conclusions and Relevance Despite increasing clinical actionability for male carriers of BRCA1/2 PVs, far fewer males than female individuals undergo cancer genetic testing. Oncologists, internists, and primary care clinicians should be vigilant about offering appropriate genetic testing to males. Identifying more male carriers of BRCA1/2 PVs will maximize opportunities for cancer early detection, targeted risk management, and cancer treatment for males, along with facilitating opportunities for risk reduction and prevention in their family members, thereby decreasing the burden of hereditary cancer.
Collapse
Affiliation(s)
- Heather H Cheng
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Medicine (Hematology and Oncology), University of Washington, Seattle
| | - Jeffrey W Shevach
- Division of Medical Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Elena Castro
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fergus J Couch
- Division of Experimental Pathology and Laboratory Medicine, Mayo Clinic, Rochester, New York
| | - Susan M Domchek
- Department of Medicine, Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Rosalind A Eeles
- The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Veda N Giri
- Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut
| | - Michael J Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Mary-Claire King
- Department of Medicine (Medical Genetics) and Department of Genome Sciences, University of Washington, Seattle
| | - Daniel W Lin
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Urology, University of Washington, Seattle
| | - Stacy Loeb
- Department of Urology and Population Health, New York University School of Medicine, New York
- Department of Surgery/Urology, Manhattan Veterans Affairs, New York, New York
| | - Todd M Morgan
- Department of Urology, University of Michigan, Ann Arbor
| | - Kenneth Offit
- Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Colin C Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle
- Brotman Baty Institute for Precision Medicine, Seattle, Washington
| | - Edward M Schaeffer
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Brittany M Szymaniak
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jason L Vassy
- Harvard Medical School at VA Boston Healthcare System, Boston, Massachusetts
| | - Bryson W Katona
- Department of Medicine, Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Kara N Maxwell
- Department of Medicine, Basser Center for BRCA and Abramson Cancer Center, University of Pennsylvania, Philadelphia
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania
| |
Collapse
|
18
|
Safizadeh F, Mandic M, Hoffmeister M, Brenner H. Reevaluating the fraction of cancer attributable to excess weight: overcoming the hidden impact of prediagnostic weight loss. Eur J Epidemiol 2024; 39:991-1003. [PMID: 39294524 PMCID: PMC11470860 DOI: 10.1007/s10654-024-01146-0] [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: 01/10/2024] [Accepted: 07/30/2024] [Indexed: 09/20/2024]
Abstract
OBJECTIVE To evaluate the magnitude of the potential underestimation of the proportion of cancer cases attributable to excess weight, known as population attributable fraction (PAF), due to potential bias from prediagnostic weight loss already present at baseline of cohort studies and to overcome it as much as possible. METHODS Data from the UK Biobank cohort participants aged 40-69 without prior cancer diagnosis were analyzed. We assessed the magnitude of associations of excess weight with the incidence of obesity-related cancers combined, and separately for gastrointestinal (GI) and other cancers. Using multivariable Cox proportional hazards models, hazard ratios (HR) and their 95% confidence intervals (CI), and PAFs for excess weight at baseline were estimated for various periods of time after weight measurements. FINDINGS Of 458,660 participants, 20,218 individuals developed obesity-related cancers during a median 11.0-year follow-up, comprising 8,460 GI, and 11,765 non-GI cancers. PAFs were much higher for cancers occurring more than four years after recruitment than for cancers occurring within the initial four years: 17.7% versus 7.2%, 21.4% versus 11.7% for GI, non-GI and all obesity-related cancers combined, respectively. With respect to total cancer (including cancers with no established relationship with excess weight), PAFs were estimated as 5.1% and 8.8% for the 0-4 and 4-14-year periods of follow-up. CONCLUSION The proportion of cancers attributable to excess weight is likely substantially larger than previously estimated based on cohort studies with short follow-up time or no or only limited exclusion of the early years of follow-up from the analyses.
Collapse
Affiliation(s)
- Fatemeh Safizadeh
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, D-69120, Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Marko Mandic
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, D-69120, Heidelberg, Germany
- Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, D-69120, Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Im Neuenheimer Feld 581, D-69120, Heidelberg, Germany.
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
19
|
Patterson L, Toledo FGS, Maitra A, Chari ST. Pancreatic Cancer-Induced Metabolic Dysregulation Syndrome: Clinical Profile, Proposed Mechanisms, and Unanswered Questions. Gastroenterology 2024:S0016-5085(24)05412-X. [PMID: 39222716 DOI: 10.1053/j.gastro.2024.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 08/02/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Affiliation(s)
- LaNisha Patterson
- Department of Pathology and Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frederico G S Toledo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anirban Maitra
- Department of Pathology and Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Suresh T Chari
- Department of Gastroenterology, Hepatology, and Nutrition, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
20
|
Bogdanski AM, Onnekink AM, Inderson A, Boekestijn B, Bonsing BA, Vasen HFA, van Hooft JE, Boonstra JJ, Mieog JSD, Wasser MNJM, Feshtali S, Potjer TP, Klatte DCF, van Leerdam ME. The Added Value of Blood Glucose Monitoring in High-Risk Individuals Undergoing Pancreatic Cancer Surveillance. Pancreas 2024; 53:e566-e572. [PMID: 38598368 DOI: 10.1097/mpa.0000000000002335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
OBJECTIVES The study aimed to investigate the added value of blood glucose monitoring in high-risk individuals (HRIs) participating in pancreatic cancer surveillance. MATERIALS AND METHODS High-risk individuals with a CDKN2A/p16 germline pathogenic variant participating in pancreatic cancer surveillance were included in this study. Multivariable logistic regression was performed to assess the relationship between new-onset diabetes (NOD) and pancreatic ductal adenocarcinoma (PDAC). To quantify the diagnostic performance of NOD as a marker for PDAC, receiver operating characteristic curve with area under the curve was computed. RESULTS In total, 220 HRIs were included between 2000 and 2019. Median age was 61 (interquartile range. 53-71) years and 62.7% of participants were female. During the study period, 26 (11.8%) HRIs developed NOD, of whom 5 (19.2%) later developed PDAC. The other 23 (82.1%) PDAC cases remained NOD-free. Multivariable analysis showed no statistically significant relationship between NOD and PDAC (odds ratio, 1.21; 95% confidence interval, 0.39-3.78) and 4 of 5 PDAC cases seemed to have NOD within 3 months before diagnosis. Furthermore, NOD did not differentiate between HRIs with and without PDAC (area under the curve, 0.54; 95% confidence interval, 0.46-0.61). CONCLUSIONS In this study, we found no added value for longitudinal glucose monitoring in CDKN2A pathogenic variant carriers participating in an imaging-based pancreatic cancer surveillance program.
Collapse
Affiliation(s)
| | | | - Akin Inderson
- From the Departments of Gastroenterology and Hepatology
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Virdee PS, Collins KK, Friedemann Smith C, Yang X, Zhu S, Roberts SE, Roberts N, Oke JL, Bankhead C, Perera R, Hobbs FDR, Nicholson BD. The Association between Blood Test Trends and Undiagnosed Cancer: A Systematic Review and Critical Appraisal. Cancers (Basel) 2024; 16:1692. [PMID: 38730644 PMCID: PMC11083147 DOI: 10.3390/cancers16091692] [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: 02/28/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Clinical guidelines include monitoring blood test abnormalities to identify patients at increased risk of undiagnosed cancer. Noting blood test changes over time may improve cancer risk stratification by considering a patient's individual baseline and important changes within the normal range. We aimed to review the published literature to understand the association between blood test trends and undiagnosed cancer. MEDLINE and EMBASE were searched until 15 May 2023 for studies assessing the association between blood test trends and undiagnosed cancer. We used descriptive summaries and narratively synthesised studies. We included 29 articles. Common blood tests were haemoglobin (24%, n = 7), C-reactive protein (17%, n = 5), and fasting blood glucose (17%, n = 5), and common cancers were pancreatic (29%, n = 8) and colorectal (17%, n = 5). Of the 30 blood tests studied, an increasing trend in eight (27%) was associated with eight cancer types, and a decreasing trend in 17 (57%) with 10 cancer types. No association was reported between trends in 11 (37%) tests and breast, bile duct, glioma, haematological combined, liver, prostate, or thyroid cancers. Our review highlights trends in blood tests that could facilitate the identification of individuals at increased risk of undiagnosed cancer. For most possible combinations of tests and cancers, there was limited or no evidence.
Collapse
Affiliation(s)
- Pradeep S. Virdee
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, University of Oxford, Woodstock Road, Oxford OX2 6GG, UK; (K.K.C.); (C.F.S.); (S.Z.); (J.L.O.); (C.B.); (R.P.); (F.R.H.); (B.D.N.)
| | - Kiana K. Collins
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, University of Oxford, Woodstock Road, Oxford OX2 6GG, UK; (K.K.C.); (C.F.S.); (S.Z.); (J.L.O.); (C.B.); (R.P.); (F.R.H.); (B.D.N.)
| | - Claire Friedemann Smith
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, University of Oxford, Woodstock Road, Oxford OX2 6GG, UK; (K.K.C.); (C.F.S.); (S.Z.); (J.L.O.); (C.B.); (R.P.); (F.R.H.); (B.D.N.)
| | - Xin Yang
- St Edmund Hall, University of Oxford, Oxford OX1 4AR, UK;
| | - Sufen Zhu
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, University of Oxford, Woodstock Road, Oxford OX2 6GG, UK; (K.K.C.); (C.F.S.); (S.Z.); (J.L.O.); (C.B.); (R.P.); (F.R.H.); (B.D.N.)
| | - Sophie E. Roberts
- Medical Sciences Division, St Peters College, University of Oxford, Oxford OX1 2DL, UK;
| | - Nia Roberts
- Bodleian Health Care Libraries, Oxford OX3 7DQ, UK;
| | - Jason L. Oke
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, University of Oxford, Woodstock Road, Oxford OX2 6GG, UK; (K.K.C.); (C.F.S.); (S.Z.); (J.L.O.); (C.B.); (R.P.); (F.R.H.); (B.D.N.)
| | - Clare Bankhead
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, University of Oxford, Woodstock Road, Oxford OX2 6GG, UK; (K.K.C.); (C.F.S.); (S.Z.); (J.L.O.); (C.B.); (R.P.); (F.R.H.); (B.D.N.)
| | - Rafael Perera
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, University of Oxford, Woodstock Road, Oxford OX2 6GG, UK; (K.K.C.); (C.F.S.); (S.Z.); (J.L.O.); (C.B.); (R.P.); (F.R.H.); (B.D.N.)
| | - FD Richard Hobbs
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, University of Oxford, Woodstock Road, Oxford OX2 6GG, UK; (K.K.C.); (C.F.S.); (S.Z.); (J.L.O.); (C.B.); (R.P.); (F.R.H.); (B.D.N.)
| | - Brian D. Nicholson
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, University of Oxford, Woodstock Road, Oxford OX2 6GG, UK; (K.K.C.); (C.F.S.); (S.Z.); (J.L.O.); (C.B.); (R.P.); (F.R.H.); (B.D.N.)
| |
Collapse
|
24
|
Scherübl H. [Early detection of sporadic pancreatic cancer]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:412-419. [PMID: 37827502 DOI: 10.1055/a-2114-9847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
The incidence of pancreatic cancer is rising. At present, pancreatic cancer is the third most common cancer-causing death in Germany, but it is expected to become the second in 2030 and finally the leading cause of cancer death in 2050. Pancreatic ductal adenocarcinoma (PC) is generally diagnosed at advanced stages, and 5-year-survival has remained poor. Early detection of sporadic PC at stage IA, however, can yield a 5-year-survival rate of about 80%. Early detection initiatives aim at identifying persons at high risk. People with new-onset diabetes at age 50 or older have attracted much interest. Novel strategies regarding how to detect sporadic PC at an early stage are being discussed.
Collapse
Affiliation(s)
- Hans Scherübl
- Klinik für Innere Medizin; Gastroenterol., GI Onkol. u. Infektiol., Vivantes Klinikum Am Urban, Berlin, Germany
- Akademisches Lehrkrankenhaus der Charité, Berlin, Germany
| |
Collapse
|
25
|
Cichosz SL, Jensen MH, Hejlesen O, Henriksen SD, Drewes AM, Olesen SS. Prediction of pancreatic cancer risk in patients with new-onset diabetes using a machine learning approach based on routine biochemical parameters. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 244:107965. [PMID: 38070389 DOI: 10.1016/j.cmpb.2023.107965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/16/2023] [Accepted: 11/30/2023] [Indexed: 01/26/2024]
Abstract
OBJECTIVE To develop a machine-learning model that can predict the risk of pancreatic ductal adenocarcinoma (PDAC) in people with new-onset diabetes (NOD). METHODS From a population-based sample of individuals with NOD aged >50 years, patients with pancreatic cancer-related diabetes (PCRD), defined as NOD followed by a PDAC diagnosis within 3 years, were included (n = 716). These PCRD patients were randomly matched in a 1:1 ratio with individuals having NOD. Data from Danish national health registries were used to develop a random forest model to distinguish PCRD from Type 2 diabetes. The model was based on age, gender, and parameters derived from feature engineering on trajectories of routine biochemical variables. Model performance was evaluated using receiver operating characteristic curves (ROC) and relative risk scores. RESULTS The most discriminative model included 20 features and achieved a ROC-AUC of 0.78 (CI:0.75-0.83). Compared to the general NOD population, the relative risk for PCRD was 20-fold increase for the 1 % of patients predicted by the model to have the highest cancer risk (3-year cancer risk of 12 % and sensitivity of 20 %). Age was the most discriminative single feature, followed by the rate of change in haemoglobin A1c and the latest plasma triglyceride level. When the prediction model was restricted to patients with PDAC diagnosed six months after diabetes diagnosis, the ROC-AUC was 0.74 (CI:0.69-0.79). CONCLUSION In a population-based setting, a machine-learning model utilising information on age, sex and trajectories of routine biochemical variables demonstrated good discriminative ability between PCRD and Type 2 diabetes.
Collapse
Affiliation(s)
- 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
| | - Asbjørn Mohr Drewes
- 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
|
26
|
Claridge H, Price CA, Ali R, Cooke EA, de Lusignan S, Harvey-Sullivan A, Hodges C, Khalaf N, O'Callaghan D, Stunt A, Thomas SA, Thomson J, Lemanska A. Determining the feasibility of calculating pancreatic cancer risk scores for people with new-onset diabetes in primary care (DEFEND PRIME): study protocol. BMJ Open 2024; 14:e079863. [PMID: 38262635 PMCID: PMC10806670 DOI: 10.1136/bmjopen-2023-079863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/04/2024] [Indexed: 01/25/2024] Open
Abstract
INTRODUCTION Worldwide, pancreatic cancer has a poor prognosis. Early diagnosis may improve survival by enabling curative treatment. Statistical and machine learning diagnostic prediction models using risk factors such as patient demographics and blood tests are being developed for clinical use to improve early diagnosis. One example is the Enriching New-onset Diabetes for Pancreatic Cancer (ENDPAC) model, which employs patients' age, blood glucose and weight changes to provide pancreatic cancer risk scores. These values are routinely collected in primary care in the UK. Primary care's central role in cancer diagnosis makes it an ideal setting to implement ENDPAC but it has yet to be used in clinical settings. This study aims to determine the feasibility of applying ENDPAC to data held by UK primary care practices. METHODS AND ANALYSIS This will be a multicentre observational study with a cohort design, determining the feasibility of applying ENDPAC in UK primary care. We will develop software to search, extract and process anonymised data from 20 primary care providers' electronic patient record management systems on participants aged 50+ years, with a glycated haemoglobin (HbA1c) test result of ≥48 mmol/mol (6.5%) and no previous abnormal HbA1c results. Software to calculate ENDPAC scores will be developed, and descriptive statistics used to summarise the cohort's demographics and assess data quality. Findings will inform the development of a future UK clinical trial to test ENDPAC's effectiveness for the early detection of pancreatic cancer. ETHICS AND DISSEMINATION This project has been reviewed by the University of Surrey University Ethics Committee and received a favourable ethical opinion (FHMS 22-23151 EGA). Study findings will be presented at scientific meetings and published in international peer-reviewed journals. Participating primary care practices, clinical leads and policy makers will be provided with summaries of the findings.
Collapse
Affiliation(s)
- Hugh Claridge
- School of Health Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
- National Physical Laboratory, Teddington, UK
| | - Claire A Price
- School of Health Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
- National Physical Laboratory, Teddington, UK
| | - Rofique Ali
- Tower Hamlets Network 1 Primary Care Network, London, UK
| | | | - Simon de Lusignan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Adam Harvey-Sullivan
- Tower Hamlets Network 1 Primary Care Network, London, UK
- Centre for Primary Care, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | | | - Natalia Khalaf
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Center for Innovations in Quality, Effectiveness, and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | | | - Ali Stunt
- Pancreatic Cancer Action, Oakhanger, Hampshire, UK
| | | | | | - Agnieszka Lemanska
- School of Health Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
- National Physical Laboratory, Teddington, UK
| |
Collapse
|
27
|
Roy RV, Means N, Rao G, Asfa S, Madka V, Dey A, Zhang Y, Choudhury M, Fung KM, Dhanasekaran DN, Friedman JE, Crawford HC, Rao CV, Bhattacharya R, Mukherjee P. Pancreatic Ubap2 deletion regulates glucose tolerance, inflammation, and protection from cerulein-induced pancreatitis. Cancer Lett 2023; 578:216455. [PMID: 37865160 PMCID: PMC10897936 DOI: 10.1016/j.canlet.2023.216455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 10/23/2023]
Abstract
Ubiquitin-binding associated protein 2 (UBAP2) is reported to promote macropinocytosis and pancreatic adenocarcinoma (PDAC) growth, however, its role in normal pancreatic function remains unknown. We addressed this knowledge gap by generating UBAP2 knockout (U2KO) mice under a pancreas-specific Cre recombinase (Pdx1-Cre). Pancreatic architecture remained intact in U2KO animals, but they demonstrated slight glucose intolerance compared to controls. Upon cerulein challenge to induce pancreatitis, U2KO animals had reduced levels of several pancreatitis-relevant cytokines, amylase and lipase in the serum, reduced tissue damage, and lessened neutrophil infiltration into the pancreatic tissue. Mechanistically, cerulein-challenged U2KO animals revealed reduced NF-κB activation compared to controls. In vitro promoter binding studies confirmed the reduction of NF-κB binding to its target molecules supporting UBAP2 as a new regulator of inflammation in pancreatitis and may be exploited as a therapeutic target in future to inhibit pancreatitis.
Collapse
Affiliation(s)
- Ram Vinod Roy
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Nicolas Means
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Geeta Rao
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sima Asfa
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Venkateshwar Madka
- Center for Cancer Prevention and Drug Development, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anindya Dey
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Yushan Zhang
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Monalisa Choudhury
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kar-Ming Fung
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Danny N Dhanasekaran
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Jacob E Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Howard C Crawford
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Health System, Detroit, MI, USA
| | - Chinthalapally V Rao
- Center for Cancer Prevention and Drug Development, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Resham Bhattacharya
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Priyabrata Mukherjee
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
28
|
Park J, Lim F, Prest M, Ferris JS, Aziz Z, Agyekum A, Wagner S, Gulati R, Hur C. Quantifying the potential benefits of early detection for pancreatic cancer through a counterfactual simulation modeling analysis. Sci Rep 2023; 13:20028. [PMID: 37973858 PMCID: PMC10654404 DOI: 10.1038/s41598-023-46751-3] [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: 08/28/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023] Open
Abstract
The benefits of cancer early detection depend on various factors, including cancer type, screening method performance, stage at diagnosis, and subsequent treatment. Although numerous studies have evaluated the effectiveness of screening interventions for identifying cancer at earlier stages, there is no quantitative analysis that studies the optimal early detection time interval that results in the greatest mortality benefit; such data could serve as a target and benchmark for cancer early detection strategies. In this study, we focus on pancreatic ductal adenocarcinoma (PDAC), a cancer known for its lack of early symptoms. Consequently, it is most often detected at late stages when the 5-year survival rate is only 3%. We developed a PDAC population model that simulates an individual patient's age and stage at diagnosis, while replicating overall US cancer incidence and mortality rates. The model includes "cancer sojourn time," serving as a proxy for the speed of cancer progression, with shorter times indicating rapid progression and longer times indicating slower progression. In our PDAC model, our hypothesis was that earlier cancer detection, potentially through a hypothetical screening intervention in the counterfactual analysis, would yield reduced mortality as compared to a no-screening group. We found that the benefits of early detection, such as increased life-years gained, are greater when the sojourn time is shorter, reaching their maximum when identification is made 4-6 years prior to clinical diagnosis (e.g., when a symptomatic diagnosis is made). However, when early detection occurs even earlier, for example 6-10 years prior to clinical diagnosis, the benefits significantly diminish for shorter sojourn time cancers, and level off for longer sojourn time cancers. Our study clarifies the potential benefits of PDAC early detection that explicitly incorporates individual patient heterogeneity in cancer progression and identifies quantitative benchmarks for future interventions.
Collapse
Affiliation(s)
- Jiheum Park
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - Francesca Lim
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Matthew Prest
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jennifer S Ferris
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Zainab Aziz
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Alice Agyekum
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Sophie Wagner
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Roman Gulati
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Chin Hur
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
29
|
Katona BW, Worthington C, Clay D, Cincotta H, Ahmad NA, Ginsberg GG, Kochman ML, Brand RE. Outcomes of the IMMray PanCan-d Test in High-Risk Individuals Undergoing Pancreatic Surveillance: Pragmatic Data and Lessons Learned. JCO Precis Oncol 2023; 7:e2300445. [PMID: 37883920 DOI: 10.1200/po.23.00445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 10/28/2023] Open
Abstract
PURPOSE An effective blood-based test for pancreatic cancer (PC) screening has remained elusive. The IMMray PanCan-d is the first commercially available blood-based test specifically designed for early detection of PC; however, outcomes from its use in clinical practice have not been reported. METHODS We performed a blinded spike-in study of 100 individuals who had an IMMray PanCan-d test, including 94 high-risk individuals (HRIs) undergoing PC surveillance and six individuals with known PC. Specimens were processed blindly following the commercial laboratory's standardized operating procedure. Positive predictive value (PPV) and negative predictive value (NPV) were calculated. RESULTS Cohort characteristics included a median age of 63 (IQR, 55-70) years, 57% female, 96% non-Hispanic White, 57% with a pathogenic variant in a PC risk gene (BRCA2 most commonly-18%), and 83% with a family history of PC. Among IMMray PanCan-d results from 94 HRIs undergoing PC surveillance, there was one positive (1%), seven borderlines (7%), 73 negatives (78%), and 13 tests not performed because of low CA19-9 expression (14%). No PC was diagnosed among these HRIs; however, there were two sub-cm pancreatic neuroendocrine tumors, seven clinically diagnosed side branch intraductal papillary mucinous neoplasms ≥1 cm, and a sub-cm solid mass with indeterminate cytology requiring close follow-up; all these individuals had negative IMMray PanCan-d tests. Of the six spiked-in PCs, four (67%) yielded a positive and two (33%) yielded a negative. With an estimated disease prevalence of 2%, the PPV and NPV are 52% and 99%, respectively, if borderline results are considered negative and 12% and 99%, respectively, if borderline tests are considered positive. CONCLUSION In clinical practice, IMMray PanCan-d has a robust NPV; however, PPV is dramatically influenced by whether borderline results are characterized as a positive or negative result.
Collapse
Affiliation(s)
- Bryson W Katona
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Christine Worthington
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Daniel Clay
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | | | - Nuzhat A Ahmad
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Gregory G Ginsberg
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Michael L Kochman
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Randall E Brand
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
30
|
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
|
31
|
Lemanska A, Andrews C, Fisher L, Bacon S, Frampton AE, Mehrkar A, Inglesby P, Davy S, Roberts K, Patalay P, Goldacre B, MacKenna B, Walker AJ. Healthcare in England was affected by the COVID-19 pandemic across the pancreatic cancer pathway: A cohort study using OpenSAFELY-TPP. eLife 2023; 12:e85332. [PMID: 37561116 PMCID: PMC10414967 DOI: 10.7554/elife.85332] [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: 12/03/2022] [Accepted: 07/14/2023] [Indexed: 08/11/2023] Open
Abstract
Background Healthcare across all sectors, in the UK and globally, was negatively affected by the COVID-19 pandemic. We analysed healthcare services delivered to people with pancreatic cancer from January 2015 to March 2023 to investigate the effect of the COVID-19 pandemic. Methods With the approval of NHS England, and drawing from a nationally representative OpenSAFELY-TPP dataset of 24 million patients (over 40% of the English population), we undertook a cohort study of people diagnosed with pancreatic cancer. We queried electronic healthcare records for information on the provision of healthcare services across the pancreatic cancer pathway. To estimate the effect of the COVID-19 pandemic, we predicted the rates of healthcare services if the pandemic had not happened. We used generalised linear models and the pre-pandemic data from January 2015 to February 2020 to predict rates in March 2020 to March 2023. The 95% confidence intervals of the predicted values were used to estimate the significance of the difference between the predicted and observed rates. Results The rate of pancreatic cancer and diabetes diagnoses in the cohort was not affected by the pandemic. There were 26,840 people diagnosed with pancreatic cancer from January 2015 to March 2023. The mean age at diagnosis was 72 (±11 SD), 48% of people were female, 95% were of White ethnicity, and 40% were diagnosed with diabetes. We found a reduction in surgical resections by 25-28% during the pandemic. In addition, 20%, 10%, and 4% fewer people received body mass index, glycated haemoglobin, and liver function tests, respectively, before they were diagnosed with pancreatic cancer. There was no impact of the pandemic on the number of people making contact with primary care, but the number of contacts increased on average by 1-2 per person amongst those who made contact. Reporting of jaundice decreased by 28%, but recovered within 12 months into the pandemic. Emergency department visits, hospital admissions, and deaths were not affected. Conclusions The pandemic affected healthcare in England across the pancreatic cancer pathway. Positive lessons could be learnt from the services that were resilient and those that recovered quickly. The reductions in healthcare experienced by people with cancer have the potential to lead to worse outcomes. Current efforts should focus on addressing the unmet needs of people with cancer. Funding This work was jointly funded by the Wellcome Trust (222097/Z/20/Z); MRC (MR/V015757/1, MC_PC-20059, MR/W016729/1); NIHR (NIHR135559, COV-LT2-0073), and Health Data Research UK (HDRUK2021.000, 2021.0157). This work was funded by Medical Research Council (MRC) grant reference MR/W021390/1 as part of the postdoctoral fellowship awarded to AL and undertaken at the Bennett Institute, University of Oxford. The views expressed are those of the authors and not necessarily those of the NIHR, NHS England, UK Health Security Agency (UKHSA), or the Department of Health and Social Care. Funders had no role in the study design, collection, analysis, and interpretation of data; in the writing of the report; and in the decision to submit the article for publication.
Collapse
Affiliation(s)
- Agnieszka Lemanska
- Faculty of Health and Medical Sciences, University of SurreyGuildfordUnited Kingdom
| | - Colm Andrews
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of OxfordOxfordUnited Kingdom
| | - Louis Fisher
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of OxfordOxfordUnited Kingdom
| | - Seb Bacon
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of OxfordOxfordUnited Kingdom
| | - Adam E Frampton
- Faculty of Health and Medical Sciences, University of SurreyGuildfordUnited Kingdom
- HPB Surgical Unit, Royal Surrey NHS Foundation TrustGuildfordUnited Kingdom
- Oncology Section, Surrey Cancer Research Institute, Department of Clinical and Experimental Medicine, University of SurreyGuildfordUnited Kingdom
| | - Amir Mehrkar
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of OxfordOxfordUnited Kingdom
| | - Peter Inglesby
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of OxfordOxfordUnited Kingdom
| | - Simon Davy
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of OxfordOxfordUnited Kingdom
| | - Keith Roberts
- Institute of Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
| | - Praveetha Patalay
- MRC Unit for Lifelong Health and Ageing and Centre for Longitudinal Studies, University College LondonLondonUnited Kingdom
| | - Ben Goldacre
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of OxfordOxfordUnited Kingdom
| | - Brian MacKenna
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of OxfordOxfordUnited Kingdom
| | - Alex J Walker
- Bennett Institute for Applied Data Science, Nuffield Department of Primary Care Health Sciences, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
32
|
White MJ, Sheka AC, LaRocca CJ, Irey RL, Ma S, Wirth KM, Benner A, Denbo JW, Jensen EH, Ankeny JS, Ikramuddin S, Tuttle TM, Hui JYC, Marmor S. The association of new-onset diabetes with subsequent diagnosis of pancreatic cancer-novel use of a large administrative database. J Public Health (Oxf) 2023; 45:e266-e274. [PMID: 36321614 PMCID: PMC10273390 DOI: 10.1093/pubmed/fdac118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 09/05/2022] [Accepted: 09/26/2022] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Screening options for pancreatic ductal adenocarcinoma (PDAC) are limited. New-onset type 2 diabetes (NoD) is associated with subsequent diagnosis of PDAC in observational studies and may afford an opportunity for PDAC screening. We evaluated this association using a large administrative database. METHODS Patients were identified using claims data from the OptumLabs® Data Warehouse. Adult patients with NoD diagnosis were matched 1:3 with patients without NoD using age, sex and chronic obstructive pulmonary disease (COPD) status. The event of PDAC diagnosis was compared between cohorts using the Kaplan-Meier method. Factors associated with PDAC diagnosis were evaluated with Cox's proportional hazards modeling. RESULTS We identified 640 421 patients with NoD and included 1 921 263 controls. At 3 years, significantly more PDAC events were identified in the NoD group vs control group (579 vs 505; P < 0.001). When controlling for patient factors, NoD was significantly associated with elevated risk of PDAC (HR 3.474, 95% CI 3.082-3.920, P < 0.001). Other factors significantly associated with PDAC diagnosis were increasing age, increasing age among Black patients, and COPD diagnosis (P ≤ 0.05). CONCLUSIONS NoD was independently associated with subsequent diagnosis of PDAC within 3 years. Future studies should evaluate the feasibility and benefit of PDAC screening in patients with NoD.
Collapse
Affiliation(s)
- M J White
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
| | - A C Sheka
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- OptumLabs® Visiting Fellow, Eden Prairie, MN, USA Institute for Health Informatics, University of Minnesota, Minneapolis MN, 55455 USA
| | - C J LaRocca
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455, USA
| | - R L Irey
- Clinical and Translational Science Institute, University of Minnesota, Minneapolis MN, 55455 USA
| | - S Ma
- Clinical and Translational Science Institute, University of Minnesota, Minneapolis MN, 55455 USA
| | - K M Wirth
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- OptumLabs® Visiting Fellow, Eden Prairie, MN, USA Institute for Health Informatics, University of Minnesota, Minneapolis MN, 55455 USA
| | - A Benner
- Clinical and Translational Science Institute, University of Minnesota, Minneapolis MN, 55455 USA
| | - J W Denbo
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa FL 33612 USA
| | - E H Jensen
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455, USA
| | - J S Ankeny
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455, USA
| | - S Ikramuddin
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- OptumLabs® Visiting Fellow, Eden Prairie, MN, USA Institute for Health Informatics, University of Minnesota, Minneapolis MN, 55455 USA
| | - T M Tuttle
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455, USA
| | - J Y C Hui
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455, USA
| | - S Marmor
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455, USA
- Clinical and Translational Science Institute, University of Minnesota, Minneapolis MN, 55455 USA
| |
Collapse
|
33
|
Choi CCM, Arafat Y, Shamassi M, Choi J. Challenges in managing upper gastrointestinal bleeding secondary to primary squamous cell carcinoma of the pancreas: a case report and literature review. Surg Case Rep 2023; 9:77. [PMID: 37171715 PMCID: PMC10182212 DOI: 10.1186/s40792-023-01663-2] [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: 02/22/2023] [Accepted: 05/08/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND Primary pancreatic squamous cell carcinoma (SCC) is a rare type of pancreatic cancer, with an incidence of 5% of all pancreatic cancers. This condition is associated with a poor prognosis, and no optimal treatment has been established (Zhang et al. in Medicine (Baltim). 97:e12253, 2018). CASE PRESENTATION A 56-year-old man presented to our hospital with upper gastrointestinal bleeding and new-onset diabetes mellitus. He had no other medical comorbidities, episodes of pancreatitis and symptoms secondary to pancreatic insufficiency. A computed tomography (CT) scan showed a 94 × 72 × 83 mm necrotic pancreatic body mass with gastric invasion and multiple liver metastases. Gastroscopy revealed deep ulcerations at the posterior wall of the stomach with an active slow ooze. Endoscopic ultrasound was performed with EUS guided biopsy, which confirmed poorly differentiated squamous carcinoma of the pancreas. The patient underwent palliative radiotherapy for recurrent upper gastrointestinal bleeding followed by palliative chemotherapy with gemcitabine and nab-paclitaxel. He was referred to dietitians and diabetes educators for the management of pancreatic exocrine and endocrine insufficiency before being referred to community palliative care upon discharge. CONCLUSIONS This is the first reported Australian case of pancreatic SCC presenting with upper gastrointestinal bleeding and new-onset diabetes mellitus. Patients with unresectable disease require a multidisciplinary approach to manage complications and improve symptom control. However, there are no standard treatment guidelines and future research is needed in this regard.
Collapse
Affiliation(s)
- Colin Chan-Min Choi
- Department of Upper Gastrointestinal/Hepatobiliary Surgery, Western Health, Footscray, VIC, Australia.
| | - Yasser Arafat
- Department of Surgery, Western Precinct, University of Melbourne, Melbourne, Australia
- Department of Colorectal and General Surgery, Western Health, Footscray, VIC, Australia
| | - Maryam Shamassi
- Department of Anatomical Pathology, Western Health (Dorevitch Pathology), Footscray, VIC, Australia
| | - Julian Choi
- Department of Upper Gastrointestinal/Hepatobiliary Surgery, Western Health, Footscray, VIC, Australia
| |
Collapse
|
34
|
Balaban DV, Coman L, Balaban M, Zoican A, Pușcașu DA, Ayatollahi S, Mihălțeanu E, Costache RS, Ioniță-Radu F, Jinga M. Glycemic Abnormalities in Pancreatic Cystic Lesions—A Single-Center Retrospective Analysis. GASTROENTEROLOGY INSIGHTS 2023; 14:191-203. [DOI: doi.org/10.3390/gastroent14020015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2023] Open
Abstract
Background and Objectives: Glucose metabolism alterations are very common in solid pancreatic lesions, particularly in pancreatic cancer. Similarly, diabetes and especially new-onset diabetes (NOD) have been associated with the malignant transformation of pancreatic cysts. We aimed to assess the prevalence and relevant associations of glycemic abnormalities in pancreatic cystic lesions (PCLs) in a retrospective analysis. Materials and Methods: We retrospectively recruited all patients who underwent endoscopic ultrasound for a PCL over a period of 36 months (January 2018 to December 2021). Final diagnosis was set by means of tissue acquisition, surgery, follow-up, or board decision. Demographic and clinical data, laboratory workup, and imaging features were extracted from the patients’ charts according to a predefined protocol. We considered fasting blood glucose (FBG) and HbA1c values and stratified the patients as nondiabetic (FBG ≤ 99 mg/dL, HbA1c ≤ 5.6%, no history of glycemic abnormalities), prediabetic (FBG 100–125 mg/dL, HbA1c 5.7–6.4%), or diabetic (long-lasting diabetes or NOD). Results: Altogether, 81 patients were included, with a median age of 66 years, and 54.3% of them were male. The overall prevalence of fasting hyperglycemia was 54.3%, comprising 34.6% prediabetes and 22.2% diabetes, of which 16.7% had NOD. The mean FBG and HbA1c levels were higher in malignant and premalignant PCLs (intraductal papillary mucinous neoplasm (IPMN), mucinous cystic neoplasm (MCN), cystadenocarcinoma, and cystic neuroendocrine tumor) compared to the benign lesions (pseudocysts, walled-off necrosis, and serous cystadenoma): 117.0 mg/dL vs. 108.3 mg/dL and 6.1% vs. 5.5%, respectively. Conclusions: Hyperglycemia and diabetes are common in PCLs, with a high prevalence in premalignant and malignant cysts. Screening and follow-up for glycemic abnormalities should be routinely conducted for PCLs, as they can contribute to a tailored risk assessment of cysts.
Collapse
Affiliation(s)
- Daniel Vasile Balaban
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Laura Coman
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Marina Balaban
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Andreea Zoican
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Danusia Adriana Pușcașu
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Simin Ayatollahi
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Emanuela Mihălțeanu
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Raluca Simona Costache
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Florentina Ioniță-Radu
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Mariana Jinga
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| |
Collapse
|
35
|
Balaban DV, Coman L, Balaban M, Zoican A, Pușcașu DA, Ayatollahi S, Mihălțeanu E, Costache RS, Ioniță-Radu F, Jinga M. Glycemic Abnormalities in Pancreatic Cystic Lesions—A Single-Center Retrospective Analysis. GASTROENTEROLOGY INSIGHTS 2023; 14:191-203. [DOI: 10.3390/gastroent14020015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Background and Objectives: Glucose metabolism alterations are very common in solid pancreatic lesions, particularly in pancreatic cancer. Similarly, diabetes and especially new-onset diabetes (NOD) have been associated with the malignant transformation of pancreatic cysts. We aimed to assess the prevalence and relevant associations of glycemic abnormalities in pancreatic cystic lesions (PCLs) in a retrospective analysis. Materials and Methods: We retrospectively recruited all patients who underwent endoscopic ultrasound for a PCL over a period of 36 months (January 2018 to December 2021). Final diagnosis was set by means of tissue acquisition, surgery, follow-up, or board decision. Demographic and clinical data, laboratory workup, and imaging features were extracted from the patients’ charts according to a predefined protocol. We considered fasting blood glucose (FBG) and HbA1c values and stratified the patients as nondiabetic (FBG ≤ 99 mg/dL, HbA1c ≤ 5.6%, no history of glycemic abnormalities), prediabetic (FBG 100–125 mg/dL, HbA1c 5.7–6.4%), or diabetic (long-lasting diabetes or NOD). Results: Altogether, 81 patients were included, with a median age of 66 years, and 54.3% of them were male. The overall prevalence of fasting hyperglycemia was 54.3%, comprising 34.6% prediabetes and 22.2% diabetes, of which 16.7% had NOD. The mean FBG and HbA1c levels were higher in malignant and premalignant PCLs (intraductal papillary mucinous neoplasm (IPMN), mucinous cystic neoplasm (MCN), cystadenocarcinoma, and cystic neuroendocrine tumor) compared to the benign lesions (pseudocysts, walled-off necrosis, and serous cystadenoma): 117.0 mg/dL vs. 108.3 mg/dL and 6.1% vs. 5.5%, respectively. Conclusions: Hyperglycemia and diabetes are common in PCLs, with a high prevalence in premalignant and malignant cysts. Screening and follow-up for glycemic abnormalities should be routinely conducted for PCLs, as they can contribute to a tailored risk assessment of cysts.
Collapse
Affiliation(s)
- Daniel Vasile Balaban
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Laura Coman
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Marina Balaban
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Andreea Zoican
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Danusia Adriana Pușcașu
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Simin Ayatollahi
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Emanuela Mihălțeanu
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Raluca Simona Costache
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Florentina Ioniță-Radu
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Mariana Jinga
- Internal Medicine and Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Gastroenterology Department, Central Military Emergency University Hospital, 010825 Bucharest, Romania
| |
Collapse
|
36
|
Zhang Y, Wang QL, Yuan C, Lee AA, Babic A, Ng K, Perez K, Nowak JA, Lagergren J, Stampfer MJ, Giovannucci EL, Sander C, Rosenthal MH, Kraft P, Wolpin BM. Pancreatic cancer is associated with medication changes prior to clinical diagnosis. Nat Commun 2023; 14:2437. [PMID: 37117188 PMCID: PMC10147931 DOI: 10.1038/s41467-023-38088-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 04/11/2023] [Indexed: 04/30/2023] Open
Abstract
Patients with pancreatic ductal adenocarcinoma (PDAC) commonly develop symptoms and signs in the 1-2 years before diagnosis that can result in changes to medications. We investigate recent medication changes and PDAC diagnosis in Nurses' Health Study (NHS; females) and Health Professionals Follow-up Study (HPFS; males), including up to 148,973 U.S. participants followed for 2,994,057 person-years and 991 incident PDAC cases. Here we show recent initiation of antidiabetic (NHS) or anticoagulant (NHS, HFS) medications and cessation of antihypertensive medications (NHS, HPFS) are associated with pancreatic cancer diagnosis in the next 2 years. Two-year PDAC risk increases as number of relevant medication changes increases (P-trend <1 × 10-5), with participants who recently start antidiabetic and stop antihypertensive medications having multivariable-adjusted hazard ratio of 4.86 (95%CI, 1.74-13.6). These changes are not associated with diagnosis of other digestive system cancers. Recent medication changes should be considered as candidate features in multi-factor risk models for PDAC, though they are not causally implicated in development of PDAC.
Collapse
Affiliation(s)
- Yin Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Qiao-Li Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Department of Clinical Science, Intervention and Technology, Karolinka Institutet, Stockholm, Sweden
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Alice A Lee
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Kimberly Perez
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jesper Lagergren
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Meir J Stampfer
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Edward L Giovannucci
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Chris Sander
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Michael H Rosenthal
- Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
37
|
Soret B, Hense J, Lüdtke S, Thale I, Schwab A, Düfer M. Pancreatic K Ca3.1 channels in health and disease. Biol Chem 2023; 404:339-353. [PMID: 36571487 DOI: 10.1515/hsz-2022-0232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/24/2022] [Indexed: 12/27/2022]
Abstract
Ion channels play an important role for regulation of the exocrine and the endocrine pancreas. This review focuses on the Ca2+-regulated K+ channel KCa3.1, encoded by the KCNN4 gene, which is present in both parts of the pancreas. In the islets of Langerhans, KCa3.1 channels are involved in the regulation of membrane potential oscillations characterizing nutrient-stimulated islet activity. Channel upregulation is induced by gluco- or lipotoxic conditions and might contribute to micro-inflammation and impaired insulin release in type 2 diabetes mellitus as well as to diabetes-associated renal and vascular complications. In the exocrine pancreas KCa3.1 channels are expressed in acinar and ductal cells. They are thought to play a role for anion secretion during digestion but their physiological role has not been fully elucidated yet. Pancreatic carcinoma, especially pancreatic ductal adenocarcinoma (PDAC), is associated with drastic overexpression of KCa3.1. For pharmacological targeting of KCa3.1 channels, we are discussing the possible benefits KCa3.1 channel inhibitors might provide in the context of diabetes mellitus and pancreatic cancer, respectively. We are also giving a perspective for the use of a fluorescently labeled derivative of the KCa3.1 blocker senicapoc as a tool to monitor channel distribution in pancreatic tissue. In summary, modulating KCa3.1 channel activity is a useful strategy for exo-and endocrine pancreatic disease but further studies are needed to evaluate its clinical suitability.
Collapse
Affiliation(s)
- Benjamin Soret
- University of Münster, Institute of Physiology II, Robert-Koch-Straße 27b, D-48149 Münster, Germany
| | - Jurek Hense
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, Corrensstraße 48, D-48149 Münster, Germany
| | - Simon Lüdtke
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, Corrensstraße 48, D-48149 Münster, Germany
| | - Insa Thale
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Corrensstraße 48, D-48149 Münster, Germany
| | - Albrecht Schwab
- University of Münster, Institute of Physiology II, Robert-Koch-Straße 27b, D-48149 Münster, Germany
| | - Martina Düfer
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, Corrensstraße 48, D-48149 Münster, Germany
| |
Collapse
|
38
|
Tovar DR, Rosenthal MH, Maitra A, Koay EJ. Potential of artificial intelligence in the risk stratification for and early detection of pancreatic cancer. ARTIFICIAL INTELLIGENCE SURGERY 2023; 3:14-26. [PMID: 37124705 PMCID: PMC10141523 DOI: 10.20517/ais.2022.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third most lethal cancer in the United States, with a 5-year life expectancy of 11%. Most symptoms manifest at an advanced stage of the disease when surgery is no longer appropriate. The dire prognosis of PDAC warrants new strategies to improve the outcomes of patients, and early detection has garnered significant attention. However, early detection of PDAC is most often incidental, emphasizing the importance of developing new early detection screening strategies. Due to the low incidence of the disease in the general population, much of the focus for screening has turned to individuals at high risk of PDAC. This enriches the screening population and balances the risks associated with pancreas interventions. The cancers that are found in these high-risk individuals by MRI and/or EUS screening show favorable 73% 5-year overall survival. Even with the emphasis on screening in enriched high-risk populations, only a minority of incident cancers are detected this way. One strategy to improve early detection outcomes is to integrate artificial intelligence (AI) into biomarker discovery and risk models. This expert review summarizes recent publications that have developed AI algorithms for the applications of risk stratification of PDAC using radiomics and electronic health records. Furthermore, this review illustrates the current uses of radiomics and biomarkers in AI for early detection of PDAC. Finally, various challenges and potential solutions are highlighted regarding the use of AI in medicine for early detection purposes.
Collapse
Affiliation(s)
- Daniela R. Tovar
- Department of Gastrointestinal Radiation Oncology, The University of Texas, Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Anirban Maitra
- Department of Radiology, The University of Texas, Anderson Cancer Center, Houston, TX 77030, USA
| | - Eugene J. Koay
- Department of Gastrointestinal Radiation Oncology, The University of Texas, Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
39
|
Yang J, Tan C, Zheng Z, Wang X, Liu X, Chen Y. Elevated Bile Acid Is Associated with Worsened Impaired Glucose Homeostasis in Pancreatic Ductal Adenocarcinoma Patients with Extrahepatic Cholestasis through Increased Hepatic Insulin Clearance. J Clin Med 2023; 12:jcm12062352. [PMID: 36983352 PMCID: PMC10052524 DOI: 10.3390/jcm12062352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/22/2023] Open
Abstract
Background: Patients after pancreaticoduodenectomy (PD) showed improved glucose tolerance. Evidence for the effect of extrahepatic cholestasis on impaired glucose homeostasis secondary to ductal adenocarcinoma of the pancreatic head is limited. Methods: In this prospective cross-sectional study, 50 patients with ductal adenocarcinoma of the pancreatic head were included to assess the effect of extrahepatic cholestasis on glucose tolerance status based on the oral glucose tolerance test (OGTT) before pancreatic surgery. Results: Patients with extrahepatic cholestasis more frequently suffered from worsened impaired glucose homeostasis (prediabetes and new-onset diabetes, 95.2% vs. 58.6%, p = 0.004). Elevated bile acid level was recognized as an independent risk factor for impaired glucose homeostasis (p = 0.024, OR = 6.85). Hepatic insulin clearance (HIC) was significantly higher in patients with elevated bile acid levels (p = 0.001). A strong positive correlation was found between bile acid levels and HIC (r = 0.45, p = 0.001). Conclusions: This study suggested a connection between elevated bile acid levels and worsened impaired glucose homeostasis through increased insulin clearance function in ductal adenocarcinoma of pancreatic head patients.
Collapse
|
40
|
Scherübl H. [Prevention of pancreatic cancer]. Dtsch Med Wochenschr 2023; 148:246-252. [PMID: 36848888 DOI: 10.1055/a-1975-2366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
The incidence of pancreatic cancer is rising in Germany. At present pancreatic cancer is the third commonest cause of cancer death but is expected to become the second in 2030 and finally the leading cause of cancer death in 2050. Pancreatic ductal adenocarcinoma (PC) is generally diagnosed at far advanced stages and 5-year-survival has remained poor. Modifiable risk factors of PC are tobacco smoking, excess body weight, alcohol use, type 2-diabetes and the metabolic syndrome. Smoking cessation and -in case of obesity- intentional weight loss can reduce PC risk by as much as 50 %. Early detection of asymptomatic sporadic PC at stage IA - stage IA-PC now has a 5-year-survival rate of about 80 %- has become a realistic chance for people older than 50 years with new-onset diabetes.
Collapse
Affiliation(s)
- Hans Scherübl
- Klinik für Gastroenterologie, Gastrointestinale Onkologie und Infektiologie, Vivantes Klinikum am Urban, 10967 Berlin
| |
Collapse
|
41
|
Zhu X, Xu H, Xiao Z, Liu H, Ni Q, Yu X, Luo G. Hyperglycemia predicts adverse prognosis in advanced pancreatic cancer patients. Endocrine 2023; 79:296-303. [PMID: 36609907 DOI: 10.1007/s12020-022-03196-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/05/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND Diabetes mellitus is a prevalent comorbidity in pancreatic cancer. Previous studies have mainly concentrated on the association between diabetes and pancreatic cancer outcomes. However, research on the impact of hyperglycemia on the prognosis of patients with advanced pancreatic cancer is limited. METHODS Information on patients with advanced pancreatic cancer was collected from a prospectively maintained database, and the patients were divided into the hyperglycemia group (fasting blood glucose ≥7.0 mmol/L) and the normoglycemia group (fasting blood glucose < 7.0 mmol/L). Patients with preexisting diabetes were not included in these groups. The associations between hyperglycemia and clinical variables or prognosis were analyzed. RESULTS Among 697 patients with advanced pancreatic cancer and no prior history of diabetes, 25.3% were diagnosed with hyperglycemia. Patients older than 65 years had a higher risk of developing hyperglycemia (P = 0.044). Patients with hyperglycemia had a worse prognosis than those with normoglycemia (median survival, 7.5 vs. 8.8 months, P < 0.001). Hyperglycemia was associated with increased mortality (hazard ratio = 1.38; P = 0.003). CONCLUSIONS Hyperglycemia predicts worse overall survival in patients with advanced pancreatic cancer.
Collapse
Affiliation(s)
- Xinzhe Zhu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Huaxiang Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zhiwen Xiao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - He Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Quanxing Ni
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Guopei Luo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
42
|
Pita-Grisanti V, Dubay K, Lahooti A, Badi N, Ueltschi O, Gumpper-Fedus K, Hsueh HY, Lahooti I, Chavez-Tomar M, Terhorst S, Knoblaugh SE, Cao L, Huang W, Coss CC, Mace TA, Choueiry F, Hinton A, Mitchell JM, Schmandt R, Grinsfelder MO, Basen-Engquist K, Cruz-Monserrate Z. Physical Activity Delays Obesity-Associated Pancreatic Ductal Adenocarcinoma in Mice and Decreases Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.03.521203. [PMID: 36711764 PMCID: PMC9881853 DOI: 10.1101/2023.01.03.521203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS Obesity is a risk factor for pancreatic ductal adenocarcinoma (PDAC), a deadly disease with limited preventive strategies. Lifestyle interventions to decrease obesity might prevent obesity-associated PDAC. Here, we examined whether decreasing obesity by increased physical activity (PA) and/or dietary changes would decrease inflammation in humans and prevent PDAC in mice. METHODS Circulating inflammatory-associated cytokines of overweight and obese subjects before and after a PA intervention were compared. PDAC pre-clinical models were exposed to PA and/or dietary interventions after obesity-associated cancer initiation. Body composition, tumor progression, growth, fibrosis, inflammation, and transcriptomic changes in the adipose tissue were evaluated. RESULTS PA decreased the levels of systemic inflammatory cytokines in overweight and obese subjects. PDAC mice on a diet-induced obesity (DIO) and PA intervention, had delayed weight gain, decreased systemic inflammation, lower grade pancreatic intraepithelial neoplasia lesions, reduced PDAC incidence, and increased anti-inflammatory signals in the adipose tissue compared to controls. PA had additional cancer prevention benefits when combined with a non-obesogenic diet after DIO. However, weight loss through PA alone or combined with a dietary intervention did not prevent tumor growth in an orthotopic PDAC model. Adipose-specific targeting of interleukin (IL)-15, an anti-inflammatory cytokine induced by PA in the adipose tissue, slowed PDAC growth. CONCLUSIONS PA alone or combined with diet-induced weight loss delayed the progression of PDAC and reduced systemic and adipose inflammatory signals. Therefore, obesity management via dietary interventions and/or PA, or modulating weight loss related pathways could prevent obesity-associated PDAC in high-risk obese individuals.
Collapse
Affiliation(s)
- Valentina Pita-Grisanti
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
- The Ohio State University Interdisciplinary Nutrition Program, The Ohio State University, Columbus, OH
| | - Kelly Dubay
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Ali Lahooti
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Niharika Badi
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Olivia Ueltschi
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Kristyn Gumpper-Fedus
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Hsiang-Yin Hsueh
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
- The Ohio State University Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH
| | - Ila Lahooti
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Myrriah Chavez-Tomar
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Samantha Terhorst
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Sue E. Knoblaugh
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH
| | - Lei Cao
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Wei Huang
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Christopher C. Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Thomas A. Mace
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Fouad Choueiry
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | - Alice Hinton
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, OH
| | - Jennifer M Mitchell
- Department of Veterinary Medicine and Surgery, UT MD Anderson Cancer Center, Houston, TX
| | - Rosemarie Schmandt
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, UT MD Anderson Cancer Center, Houston, TX
| | - Michaela Onstad Grinsfelder
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, UT MD Anderson Cancer Center, Houston, TX
| | - Karen Basen-Engquist
- Department of Behavioral Science, Center for Energy Balance, The University of Texas MD Anderson Cancer Center, UT MD Anderson Cancer Center, Houston, TX
| | - Zobeida Cruz-Monserrate
- Department of Internal Medicine, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| |
Collapse
|
43
|
Søreide K, Ismail W, Roalsø M, Ghotbi J, Zaharia C. Early Diagnosis of Pancreatic Cancer: Clinical Premonitions, Timely Precursor Detection and Increased Curative-Intent Surgery. Cancer Control 2023; 30:10732748231154711. [PMID: 36916724 PMCID: PMC9893084 DOI: 10.1177/10732748231154711] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The overall poor prognosis in pancreatic cancer is related to late clinical detection. Early diagnosis remains a considerable challenge in pancreatic cancer. Unfortunately, the onset of clinical symptoms in patients usually indicate advanced disease or presence of metastasis. ANALYSIS AND RESULTS Currently, there are no designated diagnostic or screening tests for pancreatic cancer in clinical use. Thus, identifying risk groups, preclinical risk factors or surveillance strategies to facilitate early detection is a target for ongoing research. Hereditary genetic syndromes are a obvious, but small group at risk, and warrants close surveillance as suggested by society guidelines. Screening for pancreatic cancer in asymptomatic individuals is currently associated with the risk of false positive tests and, thus, risk of harms that outweigh benefits. The promise of cancer biomarkers and use of 'omics' technology (genomic, transcriptomics, metabolomics etc.) has yet to see a clinical breakthrough. Several proposed biomarker studies for early cancer detection lack external validation or, when externally validated, have shown considerably lower accuracy than in the original data. Biopsies or tissues are often taken at the time of diagnosis in research studies, hence invalidating the value of a time-dependent lag of the biomarker to detect a pre-clinical, asymptomatic yet operable cancer. New technologies will be essential for early diagnosis, with emerging data from image-based radiomics approaches, artificial intelligence and machine learning suggesting avenues for improved detection. CONCLUSIONS Early detection may come from analytics of various body fluids (eg 'liquid biopsies' from blood or urine). In this review we present some the technological platforms that are explored for their ability to detect pancreatic cancer, some of which may eventually change the prospects and outcomes of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Kjetil Søreide
- Department of Gastrointestinal Surgery, HPB unit, 60496Stavanger University Hospital, Stavanger, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Gastrointestinal Translational Research Group, Laboratory for Molecular Medicine, 60496Stavanger University Hospital, Stavanger, Norway
| | - Warsan Ismail
- Department of Gastrointestinal Surgery, HPB unit, 60496Stavanger University Hospital, Stavanger, Norway
| | - Marcus Roalsø
- Department of Gastrointestinal Surgery, HPB unit, 60496Stavanger University Hospital, Stavanger, Norway.,Gastrointestinal Translational Research Group, Laboratory for Molecular Medicine, 60496Stavanger University Hospital, Stavanger, Norway.,Department of Quality and Health Technology, 60496University of Stavanger, Stavanger, Norway
| | - Jacob Ghotbi
- Department of Gastrointestinal Surgery, HPB unit, 60496Stavanger University Hospital, Stavanger, Norway
| | - Claudia Zaharia
- Gastrointestinal Translational Research Group, Laboratory for Molecular Medicine, 60496Stavanger University Hospital, Stavanger, Norway.,Department of Pathology, 60496Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
44
|
Koo DH, Han K, Park CY. Impact of cumulative hyperglycemic burden on the pancreatic cancer risk: A nationwide cohort study. Diabetes Res Clin Pract 2023; 195:110208. [PMID: 36513269 DOI: 10.1016/j.diabres.2022.110208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 11/23/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
AIMS We aimed to investigate how much cumulative hyperglycemia exposure increases pancreatic cancer risk. METHODS This study used the National Health Insurance Service Database of Claims and included 3,138,099 individuals who underwent four consecutive annual health screenings between 2009 and 2013. We defined hyperglycemic burden in two ways. First, the hyperglycemic burden was given a score from 0 to 4, with one point assigned for each time blood glucose was ≥100 mg/dL or the use of an antidiabetic drug. Furthermore, we performed semiquantitative scoring of a pre-diabetic (100-125; 1 point) and diabetic level (≥126; 2 points) and categorized into one of nine groups (hyperglycemic score 0-8). RESULTS During the median 6.2 years of follow-up, groups with a hyperglycemic burden of 1, 2, 3, and 4 had a 15%, 30%, 26%, and 67% increased pancreatic cancer risk compared with normal subjects. In semiquantitative analyses, individuals with a pre-diabetic glucose level on at least one occasion had a 14% increased the risk. Furthermore, individuals with a burden score of 8 had an 89% higher risk than subjects with a normal range. CONCLUSIONS The pancreatic cancer incidence increased significantly according to the hyperglycemic burden, defined as sustained hyperglycemic exposure, including pre-diabetic levels.
Collapse
Affiliation(s)
- Dong-Hoe Koo
- Division of Hematology/Oncology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, South Korea
| | - Cheol-Young Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
45
|
Falt P, Uricová D, Fejfar T, Šembera Š, Tachecí I. News in gastroenterology, hepatology and digestive endoscopy. VNITRNI LEKARSTVI 2023; 69:198-206. [PMID: 37468316 DOI: 10.36290/vnl.2023.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Gastroenterology, hepatology and digestive endoscopy are rapidly evolving disciplines with significant advances in the diagnostics and treatment in the entire gastrointestinal tract. The aim of our article was to summarize new perspectives on relevant situations in gastroenterology and hepatology like acute pancreatitis, functional dyspepsia, rational indication of proton pump inhibitors, inflammatory bowel diseases (IBD), cholestatic liver diseases, alcohol induced hepatitis, non-alcoholic fatty live disease (NAFLD) and patophysiology of bilirubin and bile acids. Digestive endoscopy represents an interventional part of gastroenterology and key recent topics are mentioned like pancreatic cancer screening, arteficial intelligence, resection of low-risk neoplastic lesions, enteroscopy techniques, cholangio- and pancreatiscopy and extraluminal expansion of endoscopy techniques by means of endoscopic submucosal and transmural dissection, endoscopic myotomy and lumen apposing stents.
Collapse
|
46
|
Ruze R, Chen Y, Xu R, Song J, Yin X, Wang C, Xu Q. Obesity, diabetes mellitus, and pancreatic carcinogenesis: Correlations, prevention, and diagnostic implications. Biochim Biophys Acta Rev Cancer 2023; 1878:188844. [PMID: 36464199 DOI: 10.1016/j.bbcan.2022.188844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/13/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022]
Abstract
The prevalence of obesity, diabetes mellitus (DM), and pancreatic cancer (PC) has been consistently increasing in the last two decades worldwide. Sharing various influential risk factors in genetics and environmental inducers in pathogenesis, the close correlations of these three diseases have been demonstrated in plenty of clinical studies using multiple parameters among different populations. On the contrary, most measures aimed to manage and treat obesity and DM effectively reduce the risk and prevent PC occurrence, yet certain drugs can inversely promote pancreatic carcinogenesis instead. Most importantly, an elevation of blood glucose with or without a reduction in body weight, along with other potential tools, may provide valuable clues for detecting PC at an early stage in patients with obesity and DM, favoring a timely intervention and prolonging survival. Herein, the epidemiological and etiological correlations among these three diseases and the supporting clinical evidence of their connections are first summarized to favor a better and more thorough understanding of obesity- and DM-related pancreatic carcinogenesis. After comparing the distinct impacts of different weight-lowering and anti-diabetic treatments on the risk of PC, the possible diagnostic implications of hyperglycemia and weight loss in PC screening are also addressed in detail.
Collapse
Affiliation(s)
- Rexiati Ruze
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, China; Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Yuan Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, China; Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, China; Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Jianlu Song
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, China; Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Xinpeng Yin
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, China; Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Chengcheng Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, China.
| | - Qiang Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, China.
| |
Collapse
|
47
|
Chen W, Butler RK, Lustigova E, Chari ST, Maitra A, Ann Rinaudo J, Wu BU. Risk Prediction of Pancreatic Cancer in Patients With Recent-onset Hyperglycemia: A Machine-learning Approach. J Clin Gastroenterol 2023; 57:103-110. [PMID: 35470312 PMCID: PMC9585151 DOI: 10.1097/mcg.0000000000001710] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/16/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND New-onset diabetes (NOD) has been suggested as an early indicator of pancreatic cancer. However, the definition of NOD by the American Diabetes Association requires 2 simultaneous or consecutive elevated glycemic measures. We aimed to apply a machine-learning approach using electronic health records to predict the risk in patients with recent-onset hyperglycemia. MATERIALS AND METHODS In this retrospective cohort study, health plan enrollees 50 to 84 years of age who had an elevated (6.5%+) glycated hemoglobin (HbA1c) tested in January 2010 to September 2018 with recent-onset hyperglycemia were identified. A total of 102 potential predictors were extracted. Ten imputation datasets were generated to handle missing data. The random survival forests approach was used to develop and validate risk models. Performance was evaluated by c -index, calibration plot, sensitivity, specificity, and positive predictive value. RESULTS The cohort consisted of 109,266 patients (mean age: 63.6 y). The 3-year incidence rate was 1.4 (95% confidence interval: 1.3-1.6)/1000 person-years of follow-up. The 3 models containing age, weight change in 1 year, HbA1c, and 1 of the 3 variables (HbA1c change in 1 y, HbA1c in the prior 6 mo, or HbA1c in the prior 18 mo) appeared most often out of the 50 training samples. The c -indexes were in the range of 0.81 to 0.82. The sensitivity, specificity, and positive predictive value in patients who had the top 20% of the predicted risks were 56% to 60%, 80%, and 2.5% to 2.6%, respectively. CONCLUSION Targeting evaluation at the point of recent hyperglycemia based on elevated HbA1c could offer an opportunity to identify pancreatic cancer early and possibly impact survival in cancer patients.
Collapse
Affiliation(s)
- Wansu Chen
- Kaiser Permanente Southern California Research and Evaluation, Pasadena, CA
| | - Rebecca K. Butler
- Kaiser Permanente Southern California Research and Evaluation, Pasadena, CA
| | - Eva Lustigova
- Kaiser Permanente Southern California Research and Evaluation, Pasadena, CA
| | - Suresh T. Chari
- Department of Gastroenterology, Hepatology and Nutrition, University of Texas MD Anderson Cancer Center
| | - Anirban Maitra
- Sheikh Ahmed Center for Pancreatic Cancer Research, University of Texas MD Anderson Cancer Center
| | - Jo Ann Rinaudo
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD
| | - Bechien U. Wu
- Center for Pancreatic Care, Department of Gastroenterology, Los Angeles Medical Center, Southern California Permanente Medical Group, Los Angeles, CA
| |
Collapse
|
48
|
Kiritani S, Ono Y, Takamatsu M, Oba A, Sato T, Ito H, Inoue Y, Takahashi Y. Diabetogenic liver metastasis from pancreatic cancer: a case report. Surg Case Rep 2022; 8:224. [PMID: 36576596 PMCID: PMC9797629 DOI: 10.1186/s40792-022-01582-8] [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: 11/04/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Although new-onset diabetes has been described in up to 20% of patients with newly diagnosed pancreatic cancer, reports regarding new-onset diabetes associated with newly developed liver metastasis from pancreatic cancer are limited. CASE PRESENTATION A 60-year-old man was diagnosed with pancreatic tail cancer without impaired glycemic control. A curative-intent distal pancreatectomy with adjuvant S-1 chemotherapy was performed. Two years after surgery, a high HbA1c concentration and solitary liver metastasis were identified on follow-up examination. Two major chemotherapy regimens, gemcitabine/nab-paclitaxel and modified FOLFIRINOX, were sequentially administered to the patient; however, his carbohydrate 19-9 concentration continued to increase. Because the patient's glycemic control rapidly worsened in synchrony with the tumor growth, insulin therapy was initiated. Although the liver metastasis was refractory to chemotherapy, curative-intent left hepatectomy was performed because only one tumor remained. His impaired glycemic control improved immediately after surgery, and insulin therapy was terminated. When writing this report (2 years after hepatectomy), the patient was alive and recurrence-free. CONCLUSIONS New-onset diabetes appeared with the progression of metachronous liver metastasis from pancreatic cancer, without recurrence at any other site. The patient's diabetic state was improved by resection of the liver tumor, and liver metastasis itself was proven to have caused the glucometabolic disorder by increasing insulin resistance.
Collapse
Affiliation(s)
- Sho Kiritani
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital of the Japanese Foundation for Clinical Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Yoshihiro Ono
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital of the Japanese Foundation for Clinical Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan.
| | - Manabu Takamatsu
- Department of Pathology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Atsushi Oba
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital of the Japanese Foundation for Clinical Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Takafumi Sato
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital of the Japanese Foundation for Clinical Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Hiromichi Ito
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital of the Japanese Foundation for Clinical Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Yosuke Inoue
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital of the Japanese Foundation for Clinical Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| | - Yu Takahashi
- Division of Hepatobiliary and Pancreatic Surgery, Cancer Institute Hospital of the Japanese Foundation for Clinical Research, 3-8-31 Ariake, Koto-Ku, Tokyo, 135-8550, Japan
| |
Collapse
|
49
|
Lee HS, Chae W, Sung MJ, Keum J, Jo JH, Chung MJ, Park JY, Park SW, Song SY, Park EC, Nam CM, Jang SI, Bang S. Difference of risk of pancreatic cancer in new-onset diabetes and long-standing diabetes: population-based cohort study. J Clin Endocrinol Metab 2022; 108:1338-1347. [PMID: 36548964 DOI: 10.1210/clinem/dgac728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
CONTEXT Considering the absence of methods to find pancreatic cancer early, surveillance of high-risk groups is needed for early diagnosis. OBJECTIVE The study aimed to investigate the effect in the incidence of pancreatic cancer and the differences between new-onset DM (NODM) and long-standing DM (LSDM) since NODM group is a representative high-risk group. METHODS The Korean National Health Insurance Service-National Sample Cohort between 2002 and 2013 data was used. Regarding 88,396 people with DM (case group), we conducted a 1:1 propensity score matching to select a matched non-DM population (control group). To investigate the interaction between DM and the time variable distinguishing NODM and LSDM, we performed a multi-variable time-dependent Cox regression analysis. RESULTS The incidence of pancreatic cancer was higher in the DM group compared to the non-DM group (0.52% vs. 0.16%, P < 0.001). The DM group had shown different risk of pancreatic cancer development according to the duration since the DM diagnosis (NODM hazard ratio (HR): 3.81, 95% confidence interval (CI): 2.97-4.88, P < 0.001; LSDM HR: 1.53, 95% CI: 1.11-2.11, P < 0.001). When the NODM and the LSDM groups were compared, the risk of pancreatic cancer was higher in the NODM group than LSDM group (HR: 1.55, P = 0.020). In subgroup analysis, NODM group showed that men (HR = 4.42 95% CI: 3.15-6.19, P < 0.001) and patients who were in their 50 s (HR = 7.54, 95% CI: 3.24-17.56, P < 0.001) were at a higher risk of developing pancreatic cancer than matched same sex or age control group (non-DM population), respectively. CONCLUSION The risk of pancreatic cancer was greater in people with DM than non-DM population. Among people with DM, NODM showed a higher risk of pancreatic cancer than long standing DM.
Collapse
Affiliation(s)
- Hee Seung Lee
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Wonjeong Chae
- Department of Health Policy and Management, Yonsei University Graduate School of Public Health, Seoul, Republic of Korea
- Institute of Health Services Research, Yonsei University, Seoul, Republic of Korea
| | - Min Je Sung
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jiyoung Keum
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung Hyun Jo
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Moon Jae Chung
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Woo Park
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Si Young Song
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun-Cheol Park
- Institute of Health Services Research, Yonsei University, Seoul, Republic of Korea
- Department of Preventive Medicine, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Chung Mo Nam
- Department of Preventive Medicine, College of Medicine, Yonsei University, Seoul, Republic of Korea
- Department of Biostatics, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Sung-In Jang
- Institute of Health Services Research, Yonsei University, Seoul, Republic of Korea
- Department of Preventive Medicine, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Seungmin Bang
- Division of Gastroenterology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
50
|
Dong M, Cao L, Cui R, Xie Y. The connection between innervation and metabolic rearrangements in pancreatic cancer through serine. Front Oncol 2022; 12:992927. [PMID: 36582785 PMCID: PMC9793709 DOI: 10.3389/fonc.2022.992927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/31/2022] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is a kind of aggressive tumor famous for its lethality and intractability, and pancreatic ductal adenocarcinoma is the most common type. Patients with pancreatic cancer often suffer a rapid loss of weight and abdominal neuropathic pain in their early stages and then go through cachexia in the advanced stage. These features of patients are considered to be related to metabolic reprogramming of pancreatic cancer and abundant nerve innervation responsible for the pain. With increasing literature certifying the relationship between nerves and pancreatic ductal adenocarcinoma (PDAC), more evidence point out that innervation's role is not limited to neuropathic pain but explore its anti/pro-tumor functions in PDAC, especially the neural-metabolic crosstalks. This review aims to unite pancreatic cancer's innervation and metabolic rearrangements with terminated published articles. Hopefully, this article could explore the pathogenesis of PDAC and further promote promising detecting or therapeutic measurements for PDAC according to the lavish innervation in PDAC.
Collapse
Affiliation(s)
- Mengmeng Dong
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Lidong Cao
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, China,Jilin Engineering Laboratory for Translational Medicine of Hepatobiliary and Pancreatic Diseases, Second Hospital of Jilin University, Changchun, China,Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial Peoples Hospital, Hangzhou, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China,*Correspondence: Ranji Cui, ; Yingjun Xie,
| | - Yingjun Xie
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, China,Jilin Engineering Laboratory for Translational Medicine of Hepatobiliary and Pancreatic Diseases, Second Hospital of Jilin University, Changchun, China,*Correspondence: Ranji Cui, ; Yingjun Xie,
| |
Collapse
|