1
|
Yan B, Fritsche AK, Haußner E, Inamdar TV, Laumen H, Boettcher M, Gericke M, Michl P, Rosendahl J. From Genes to Environment: Elucidating Pancreatic Carcinogenesis Through Genetically Engineered and Risk Factor-Integrated Mouse Models. Cancers (Basel) 2025; 17:1676. [PMID: 40427173 PMCID: PMC12110317 DOI: 10.3390/cancers17101676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic cancer is characterized by late diagnosis, therapy resistance, and poor prognosis, necessitating the exploration of early carcinogenesis and prevention methods. Preclinical mouse models have evolved from cell line-based to human tumor tissue- or organoid-derived xenografts, now to humanized mouse models and genetically engineered mouse models (GEMMs). GEMMs, primarily driven by oncogenic Kras mutations and tumor suppressor gene alterations, offer a realistic platform for investigating pancreatic cancer initiation, progression, and metastasis. The incorporation of inducible somatic mutations and CRISPR-Cas9 screening methods has expanded their utility. To better recapitulate tumor initiation triggered by inflammatory cues, common pancreatic risk factors are being integrated into model designs. This approach aims to decipher the role of environmental factors as secondary or parallel triggers of tumor initiation alongside oncogenic burdens. Emerging models exploring pancreatitis, obesity, diabetes, and other risk factors offer significant translational potential. This review describes current mouse models for studying pancreatic carcinogenesis, their combination with inflammatory factors, and their utility in evaluating pathogenesis, providing guidance for selecting the most suitable models for pancreatic cancer research.
Collapse
Affiliation(s)
- Bin Yan
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Anne-Kristin Fritsche
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany;
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany;
| | - Erik Haußner
- Institute of Molecular Medicine, Section for Molecular Medicine of Signal Transduction, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (E.H.); (M.B.)
| | - Tanvi Vikrant Inamdar
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| | - Helmut Laumen
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| | - Michael Boettcher
- Institute of Molecular Medicine, Section for Molecular Medicine of Signal Transduction, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (E.H.); (M.B.)
| | - Martin Gericke
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany;
| | - Patrick Michl
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany; (T.V.I.); (H.L.)
| |
Collapse
|
2
|
Hu ZY, Ding D, Song Y, Deng YF, Zhang CM, Yu T. Molecular mechanism of pancreatic ductal adenocarcinoma: The heterogeneity of cancer-associated fibroblasts and key signaling pathways. World J Clin Oncol 2025; 16:97007. [PMID: 39995552 PMCID: PMC11686552 DOI: 10.5306/wjco.v16.i2.97007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/04/2024] [Accepted: 11/04/2024] [Indexed: 12/11/2024] Open
Abstract
Pancreatic ductal adenocarcinoma stands out as an exceptionally fatal cancer owing to the complexities associated with its treatment and diagnosis, leading to a notably low five-year survival rate. This study offers a detailed exploration of epidemiological trends in pancreatic cancer and key molecular drivers, such as mutations in CDKN2A, KRAS, SMAD4, and TP53, along with the influence of cancer-associated fibroblasts (CAFs) on disease progression. In particular, we focused on the pivotal roles of signaling pathways such as the transforming growth factor-β and Wnt/β-catenin pathways in the development of pancreatic cancer and investigated their application in emerging therapeutic strategies. This study provides new scientific perspectives on pancreatic cancer treatment, especially in the development of precision medicine and targeted therapeutic strategies, and demonstrates the importance of signaling pathway research in the development of effective therapeutic regimens. Future studies should explore the subtypes of CAFs and their specific roles in the tumor microenvironment to devise more effective therapeutic methods.
Collapse
Affiliation(s)
- Zhong-Yuan Hu
- First School of Clinical Medicine, Shaanxi University of Chinese Medicine, Xianyang 712000, Shaanxi Province, China
| | - Ding Ding
- First School of Clinical Medicine, Shaanxi University of Chinese Medicine, Xianyang 712000, Shaanxi Province, China
| | - Yu Song
- College of Acupuncture and Massage, Shaanxi University of Chinese Medicine, Xianyang 712000, Shaanxi Province, China
| | - Ya-Feng Deng
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou 510000, Guangdong Province, China
| | - Cheng-Ming Zhang
- Digestive Department I, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi’an 710000, Shaanxi Province, China
| | - Tao Yu
- Digestive Department I, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi’an 710000, Shaanxi Province, China
| |
Collapse
|
3
|
Sheng T, Dokmanovich B, Yu Y, Liu C, Fan H, Prizment AE, Anderson KE, Zhang J. Associations Between Intakes of Carotenoids and Tocopherols with Risk of Pancreatic Cancer in a Population-Based Case-Control Study. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2025; 44:128-136. [PMID: 39302853 PMCID: PMC11757089 DOI: 10.1080/27697061.2024.2404578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVE Experimental studies suggest that carotenoids and tocopherols modulate pancreatic carcinogenesis because they have antioxidant and other functions. We investigated the associations between intakes of these compounds and the risk of pancreatic cancer in a case-control study conducted in 1994-1998. METHODS The present analysis included 150 cases of pancreatic cancer recruited from all hospitals in the metropolitan area of the Twin Cities and Mayo Clinic and 459 controls randomly selected from the general population and frequency matched to cases by age, sex, and race. The intakes of carotenoids and tocopherols were assessed with a validated food frequency questionnaire. Unconditional logistic regression analysis was performed to evaluate the associations of interest. RESULTS The energy-adjusted intake of lutein/zeaxanthin was significantly lower in cases (2410 µg/day) than in controls (3020 µg/day). After adjustment for confounders, persons in the fourth quartile of lutein/zeaxanthin intake had a reduced risk of pancreatic cancer compared with those in the first quartile [odds ratio (OR) (95% CI): 0.40 (0.17-0.91)]. There were no significant associations with intakes of other carotenoids and tocopherols considered and with a composite score created from all individual carotenoids examined. We did not detect any significant interactions of intakes of carotenoids and tocopherols with age, sex, cigarette smoking, or alcohol intake in relation to pancreatic cancer risk. CONCLUSION The present study suggests an inverse association between lutein/zeaxanthin intake and pancreatic cancer risk, but a potential beneficial effect was not observed for other carotenoids and tocopherols.
Collapse
Affiliation(s)
- Tianchen Sheng
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
| | - Bronson Dokmanovich
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
| | - Yunpeng Yu
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
- Department of Biostatistics and Health Data Science, Indiana University Richard M. Fairbanks School of Public Health and School of Medicine, Indianapolis, IN, USA
| | - Chunliang Liu
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
- Department of Gastroenterology, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Hao Fan
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health, Bloomington, IN, USA
| | - Anna E. Prizment
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Kristin E. Anderson
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Jianjun Zhang
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, IN, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| |
Collapse
|
4
|
Grigorescu RR, Husar-Sburlan IA, Gheorghe C. Pancreatic Cancer: A Review of Risk Factors. Life (Basel) 2024; 14:980. [PMID: 39202722 PMCID: PMC11355429 DOI: 10.3390/life14080980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Pancreatic adenocarcinoma is one of the most lethal types of gastrointestinal cancer despite the latest medical advances. Its incidence has continuously increased in recent years in developed countries. The location of the pancreas can result in the initial symptoms of neoplasia being overlooked, which can lead to a delayed diagnosis and a subsequent reduction in the spectrum of available therapeutic options. The role of modifiable risk factors in pancreatic cancer has been extensively studied in recent years, with smoking and alcohol consumption identified as key contributors. However, the few screening programs that have been developed focus exclusively on genetic factors, without considering the potential impact of modifiable factors on disease occurrence. Thus, fully understanding and detecting the risk factors for pancreatic cancer represents an important step in the prevention and early diagnosis of this type of neoplasia. This review reports the available evidence on different risk factors and identifies the areas that could benefit the most from additional studies.
Collapse
Affiliation(s)
- Raluca Roxana Grigorescu
- Gastroenterology Department, “Sfanta Maria” Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | | | - Cristian Gheorghe
- Center for Digestive Disease and Liver Transplantation, Fundeni Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
5
|
Pliszka M, Szablewski L. Associations between Diabetes Mellitus and Selected Cancers. Int J Mol Sci 2024; 25:7476. [PMID: 39000583 PMCID: PMC11242587 DOI: 10.3390/ijms25137476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Cancer is one of the major causes of mortality and is the second leading cause of death. Diabetes mellitus is a serious and growing problem worldwide, and its prevalence continues to grow; it is the 12th leading cause of death. An association between diabetes mellitus and cancer has been suggested for more than 100 years. Diabetes is a common disease diagnosed among patients with cancer, and evidence indicates that approximately 8-18% of patients with cancer have diabetes, with investigations suggesting an association between diabetes and some particular cancers, increasing the risk for developing cancers such as pancreatic, liver, colon, breast, stomach, and a few others. Breast and colorectal cancers have increased from 20% to 30% and there is a 97% increased risk of intrahepatic cholangiocarcinoma or endometrial cancer. On the other hand, a number of cancers and cancer therapies increase the risk of diabetes mellitus. Complications due to diabetes in patients with cancer may influence the choice of cancer therapy. Unfortunately, the mechanisms of the associations between diabetes mellitus and cancer are still unknown. The aim of this review is to summarize the association of diabetes mellitus with selected cancers and update the evidence on the underlying mechanisms of this association.
Collapse
Affiliation(s)
- Monika Pliszka
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| | - Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| |
Collapse
|
6
|
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
|
7
|
Ali S, Na R, Tuesley K, Spilsbury K, Stewart LM, Coory M, Webb PM, Donovan P, Pearson SA, Jordan SJ, Neale RE. The association between diabetes mellitus of different durations and risk of pancreatic cancer: an Australian national data-linkage study in women. Cancer Epidemiol 2022; 81:102266. [PMID: 36240705 DOI: 10.1016/j.canep.2022.102266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/29/2022] [Accepted: 09/19/2022] [Indexed: 11/02/2022]
Abstract
AIMS The bidirectional association between diabetes mellitus (DM) and pancreatic cancer (PC) is established; however, the strength of association between duration of DM and risk of PC needs further investigation. METHODS We conducted a case-control study nested within a population-based cohort of Australian women established using record linkage. Women diagnosed with PC from July 2007 to December 2013, were matched to five controls based on age and state of residence. DM was defined according to prescription of anti-diabetic medication from administrative prescription data. We used conditional logistic regression to calculate odds ratios (OR) and 95% confidence intervals (CI), adjusted for area-level socioeconomic status, rurality of residence, weighted comorbidity score, and predicted probability of obesity. RESULTS The analyses included 7,267 cases and 35,978 controls. The mean age at the time of DM diagnosis was 71 years whereas the mean age at the time of diagnosis of PC was 76 years. A history of DM of any duration was associated with a 2-fold increase in risk of PC (OR=2.12; 95%CI:1.96-2.29) compared to having no history of DM. The risk decreased with increasing duration of DM. The highest risk was in those who had recent-onset DM (OR=8.08; 95%CI:6.88-9.50 for <12 months of DM), but the risk remained elevated with ≥5 years of DM (OR=1.40; 95%CI:1.27-1.55). CONCLUSION The markedly increased risk of PC in those with recent-onset DM emphasises the need for further research to distinguish patients for whom new-onset DM is a manifestation of PC from those with type-2 DM. The elevated risk associated with long-standing DM suggests that preventing DM may contribute to a reduction in the incidence of PC.
Collapse
Affiliation(s)
- Sitwat Ali
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; School of Public Health, University of Queensland, Brisbane, Queensland, Australia
| | - Renhua Na
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Karen Tuesley
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; School of Public Health, University of Queensland, Brisbane, Queensland, Australia
| | - Katrina Spilsbury
- Centre for Health Research, University of Notre Dame Australia, Fremantle, Western Australia, Australia
| | - Louise M Stewart
- School of Population and Global Health, The University of Western Australia, Crawley, Western Australia, Australia
| | - Michael Coory
- Centre of Research Excellence in Stillbirth, Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Penelope M Webb
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; School of Public Health, University of Queensland, Brisbane, Queensland, Australia
| | - Peter Donovan
- Royal Brisbane and Women's Hospital, Australia; Faculty of Medicine, The University of Queensland, Australia
| | - Sallie-Anne Pearson
- Centre for Big Data Research in Health, University of New South Wales UNSW, Sydney, New South Wales, Australia
| | - Susan J Jordan
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; School of Public Health, University of Queensland, Brisbane, Queensland, Australia
| | - Rachel E Neale
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; School of Public Health, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
8
|
Afghani E, Klein AP. Pancreatic Adenocarcinoma: Trends in Epidemiology, Risk Factors, and Outcomes. Hematol Oncol Clin North Am 2022; 36:879-895. [PMID: 36154788 PMCID: PMC10548451 DOI: 10.1016/j.hoc.2022.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Pancreatic cancer is one of the most lethal cancers in the world; it is a silent disease in which symptoms do not present until advanced stages, thereby reducing the 5-year survival rate to 10%. The global burden of pancreatic cancer has doubled over the past 25 years despite advancements in medicine. This review aims to discuss the global trends and disparities in pancreatic cancer, as well as the up-to-date literature on the known risk factors. A better understanding of these risk factors will reduce mortality by providing opportunities to screen these patients as well as counseling on lifestyle modifications.
Collapse
Affiliation(s)
- Elham Afghani
- Johns Hopkins School of Medicine, 1830 E Monument Street, Room 436, Baltimore, MD 21205, USA
| | - Alison P Klein
- Johns Hopkins School of Medicine, 1830 E Monument Street, Room 436, Baltimore, MD 21205, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Johns Hopkins Bloomberg School of Public Health, 1550 Orleans Street, Baltimore, MD 21231, USA.
| |
Collapse
|
9
|
Wu H, Norton V, Cui K, Zhu B, Bhattacharjee S, Lu YW, Wang B, Shan D, Wong S, Dong Y, Chan SL, Cowan D, Xu J, Bielenberg DR, Zhou C, Chen H. Diabetes and Its Cardiovascular Complications: Comprehensive Network and Systematic Analyses. Front Cardiovasc Med 2022; 9:841928. [PMID: 35252405 PMCID: PMC8891533 DOI: 10.3389/fcvm.2022.841928] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/18/2022] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus is a worldwide health problem that usually comes with severe complications. There is no cure for diabetes yet and the threat of these complications is what keeps researchers investigating mechanisms and treatments for diabetes mellitus. Due to advancements in genomics, epigenomics, proteomics, and single-cell multiomics research, considerable progress has been made toward understanding the mechanisms of diabetes mellitus. In addition, investigation of the association between diabetes and other physiological systems revealed potentially novel pathways and targets involved in the initiation and progress of diabetes. This review focuses on current advancements in studying the mechanisms of diabetes by using genomic, epigenomic, proteomic, and single-cell multiomic analysis methods. It will also focus on recent findings pertaining to the relationship between diabetes and other biological processes, and new findings on the contribution of diabetes to several pathological conditions.
Collapse
Affiliation(s)
- Hao Wu
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Vikram Norton
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Kui Cui
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Bo Zhu
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Sudarshan Bhattacharjee
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Yao Wei Lu
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Beibei Wang
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Dan Shan
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Scott Wong
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Yunzhou Dong
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Siu-Lung Chan
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Douglas Cowan
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Jian Xu
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Diane R Bielenberg
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Hong Chen
- Department of Surgery, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
10
|
Menini S, Iacobini C, Vitale M, Pesce C, Pugliese G. Diabetes and Pancreatic Cancer-A Dangerous Liaison Relying on Carbonyl Stress. Cancers (Basel) 2021; 13:313. [PMID: 33467038 PMCID: PMC7830544 DOI: 10.3390/cancers13020313] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/09/2021] [Accepted: 01/14/2021] [Indexed: 02/06/2023] Open
Abstract
Both type 2 (T2DM) and type 1 (T1DM) diabetes mellitus confer an increased risk of pancreatic cancer in humans. The magnitude and temporal trajectory of the risk conferred by the two forms of diabetes are similar, suggesting a common mechanism. Carbonyl stress is a hallmark of hyperglycemia and dyslipidemia, which accompanies T2DM, prediabetes, and obesity. Accumulating evidence demonstrates that diabetes promotes pancreatic ductal adenocarcinoma (PDAC) in experimental models of T2DM, a finding recently confirmed in a T1DM model. The carbonyl stress markers advanced glycation end-products (AGEs), the levels of which are increased in diabetes, were shown to markedly accelerate tumor development in a mouse model of Kras-driven PDAC. Consistently, inhibition of AGE formation by trapping their carbonyl precursors (i.e., reactive carbonyl species, RCS) prevented the PDAC-promoting effect of diabetes. Considering the growing attention on carbonyl stress in the onset and progression of several cancers, including breast, lung and colorectal cancer, this review discusses the mechanisms by which glucose and lipid imbalances induce a status of carbonyl stress, the oncogenic pathways activated by AGEs and their precursors RCS, and the potential use of carbonyl-scavenging agents and AGE inhibitors in PDAC prevention and treatment, particularly in high-risk diabetic individuals.
Collapse
Affiliation(s)
- Stefano Menini
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (S.M.); (C.I.); (M.V.)
| | - Carla Iacobini
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (S.M.); (C.I.); (M.V.)
| | - Martina Vitale
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (S.M.); (C.I.); (M.V.)
| | - Carlo Pesce
- Department of Neurosciences, Rehabilitation, Ophtalmology, Genetic and Maternal Infantile Sciences (DINOGMI), Department of Excellence of MIUR, University of Genoa Medical School, 16132 Genoa, Italy;
| | - Giuseppe Pugliese
- Department of Clinical and Molecular Medicine, “La Sapienza” University, 00189 Rome, Italy; (S.M.); (C.I.); (M.V.)
| |
Collapse
|
11
|
Assessment of Weight Loss and Gastrointestinal Symptoms Suggestive of Exocrine Pancreatic Dysfunction After Acute Pancreatitis. Clin Transl Gastroenterol 2020; 11:e00283. [PMID: 33464001 PMCID: PMC7743841 DOI: 10.14309/ctg.0000000000000283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Studies evaluating the natural history of exocrine pancreatic dysfunction (EPD) after acute pancreatitis (AP) are sparse. This study aims to assess incidence and predictors of weight loss and gastrointestinal (GI) symptoms suggestive of EPD 12 months after an AP episode. METHODS Patients enrolled in the Pancreatitis-associated Risk of Organ Failure Study at the time of an AP episode were included. Weight and GI symptom data were prospectively collected by self-report at enrollment and at 3- and 12-month (windows 2-7 and 8-20) telephone follow-ups. Multivariable logistic regression was used to assess factors associated with ≥10% total body weight loss (EPD surrogate) at 12 months. A generalized estimating equation was used to measure each factor's population effect (in pounds) over 12 months after AP. RESULTS Follow-up at 12 months in 186 patients (median age = 54 years, 46% men, 45% biliary, 65% first AP attack) revealed weight loss ≥10% from baseline, occurring in 44 patients (24%). Risk of weight loss increased with higher baseline body mass index, previous diagnosis of diabetes mellitus, and worsening AP severity (all P < 0.010). GI symptoms were reported in 13/31 (42%) patients at 12 months. AP severity was independently associated with ≥10% weight loss at 12 months. Over 12 months, men lost more weight than women (average 9.5 lbs); patients with severe AP lost, on average, 14 lbs. DISCUSSION Weight loss after AP occurs in one-quarter of patients and is associated with AP severity. EPD incidence after AP is likely underappreciated. Further work is needed to assess EPD and potential for pancreatic enzyme supplementation.
Collapse
|
12
|
Wang G, Xu Z, Zhu J, Ren J, Chen M, He G, Yu B. Decreased Risk in the Pancreatic Cancer With History of Hay Fever: A Meta-Analysis. Front Public Health 2020; 8:551490. [PMID: 33117769 PMCID: PMC7574341 DOI: 10.3389/fpubh.2020.551490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/21/2020] [Indexed: 01/11/2023] Open
Abstract
Background: An increasing incidence of pancreatic cancer has been observed worldwide over the last few decades. Previous reports suggested that hay fever, a common allergic disease, may function in pancreatic cancer. Data on hay fever as a risk or protective factor for pancreatic cancer was controversial in several case–control reports. So, we here did a meta-analysis on published studies to evaluate the association of hay fever and the risk of pancreatic cancer. Methods: A comprehensive literature search was performed through public databases. The association between hay fever and pancreatic cancer was evaluated by odds ratios (ORs) and 95% confidence intervals (CIs). The Cochran's Q test and I2 index were used to evaluate heterogeneity. Results: We included 8 population-based case–control studies involving 10,454 participants from 1986 to 2014. A history of hay fever was associated with a decreased risk of pancreatic cancer (OR, 0.57; 95% CI, 0.50–0.64, P < 0.00001) through fixed effect model. Conclusion: The result of our study suggested that hay fever may significantly decrease the risk of pancreatic cancer.
Collapse
Affiliation(s)
- Guannan Wang
- Blood Purification Center, Ningbo Medical Center, Lihuili Hospital, Ningbo, China
| | - Zhiwei Xu
- Department of Critical Care Medicine, Ningbo Medical Center, Lihuili Hospital, Ningbo, China
| | - Jie Zhu
- Department of Hepato-Biliary-Pancreatic Surgery, Ningbo Medical Center, Lihuili Hospital, Ningbo, China
| | - Jiayu Ren
- Department of Cardiology, Ningbo Medical Center, Lihuili Hospital, Ningbo, China
| | - Mina Chen
- Department of Endocrinology and Respiratory Medicine, Ningbo Medical Center, Lihuili Hospital, Ningbo, China
| | - Guijuan He
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou, China
| | - Beibei Yu
- Outpatient Dressing Room, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Xiangshan, China
| |
Collapse
|
13
|
Rosato V, Negri E, Bosetti C, Malats N, Gomez-Rubio P, Consortium P, Maisonneuve P, Miller AB, Bueno-de-Mesquita HB, Baghurst PA, Zatonski W, Petersen GM, Scelo G, Holcatova I, Fabianova E, Serraino D, Olson SH, Vioque J, Lagiou P, Duell EJ, Boffetta P, La Vecchia C. Gallbladder disease, cholecystectomy, and pancreatic cancer risk in the International Pancreatic Cancer Case-Control Consortium (PanC4). Eur J Cancer Prev 2020; 29:408-415. [PMID: 32740166 DOI: 10.1097/cej.0000000000000572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The association among gallbladder disease, cholecystectomy, and pancreatic cancer is unclear. Moreover, time interval between gallbladder disease or cholecystectomy and pancreatic cancer diagnosis is not considered in most previous studies. AIM To quantify the association among gallbladder disease, cholecystectomy, and pancreatic cancer, considering time since first diagnosis of gallbladder disease or cholecystectomy. METHODS We used data from nine case-control studies within the Pancreatic Cancer Case-Control Consortium, including 5760 cases of adenocarcinoma of the exocrine pancreas and 8437 controls. We estimated pooled odds ratios and the corresponding 95% confidence intervals by estimating study-specific odds ratios through multivariable unconditional logistic regression models, and then pooling the obtained estimates using fixed-effects models. RESULTS Compared with patients with no history of gallbladder disease, the pooled odds ratio of pancreatic cancer was 1.69 (95% confidence interval, 1.51-1.88) for patients reporting a history of gallbladder disease. The odds ratio was 4.90 (95% confidence interval, 3.45-6.97) for gallbladder disease diagnosed <2 years before pancreatic cancer diagnosis and 1.11 (95% confidence interval, 0.96-1.29) when ≥2 years elapsed. The pooled odds ratio was 1.64 (95% confidence interval, 1.43-1.89) for patients who underwent cholecystectomy, as compared to those without cholecystectomy. The odds ratio was 7.00 (95% confidence interval, 4.13-11.86) for a surgery <2 years before pancreatic cancer diagnosis and 1.28 (95% confidence interval, 1.08-1.53) for a surgery ≥2 years before. CONCLUSIONS There appears to be no long-term effect of gallbladder disease on pancreatic cancer risk, and at most a modest one for cholecystectomy. The strong short-term association can be explained by diagnostic bias and reverse causation.
Collapse
Affiliation(s)
- Valentina Rosato
- Unit of Medical Statistics and Biometry, National Cancer Institute, IRCCS Foundation
| | - Eva Negri
- Department of Biomedical and Clinical Sciences, University of Milan
| | - Cristina Bosetti
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO) and CIBERONC, Madrid, Spain
| | - Paulina Gomez-Rubio
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO) and CIBERONC, Madrid, Spain
| | - PanGenEU Consortium
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO) and CIBERONC, Madrid, Spain
| | - Patrick Maisonneuve
- Division of Epidemiology and Biostatistics, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Anthony B Miller
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - H Bas Bueno-de-Mesquita
- National Institute for Public Health and the Environment (RIVM), Bilthoven
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Peter A Baghurst
- Public Health, Women's and Children's Hospital, Adelaide, South Australia, Australia
| | - Witold Zatonski
- Health Promotion Foundation, Nadarzyn
- Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Gloria M Petersen
- Department of Health Sciences Research, Medicine and Medical Genetics, Mayo Clinic, Rochester, New York, USA
| | - Ghislaine Scelo
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Ivana Holcatova
- Institute of Hygiene and Epidemiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Eleonora Fabianova
- Regional Authority of Public Health, Banská Bystrica, Slovak Republic
- Faculty of Health, Catholic University, Ružomberok, Slovak Republic
| | - Diego Serraino
- Cancer Epidemiology Unit, National Cancer Institute Centro di Riferimento Oncologico, IRCCS, Aviano, Italy
| | - Sara H Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jesús Vioque
- Institute for Health and Biomedical Research ISABIAL-UMH, Alicante
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Pagona Lagiou
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, University of Athens, Athens, Greece
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Eric J Duell
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Paolo Boffetta
- The Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York, USA
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
14
|
Menini S, Iacobini C, de Latouliere L, Manni I, Vitale M, Pilozzi E, Pesce C, Cappello P, Novelli F, Piaggio G, Pugliese G. Diabetes promotes invasive pancreatic cancer by increasing systemic and tumour carbonyl stress in Kras G12D/+ mice. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:152. [PMID: 32778157 PMCID: PMC7418209 DOI: 10.1186/s13046-020-01665-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 08/04/2020] [Indexed: 01/13/2023]
Abstract
Background Type 1 and 2 diabetes confer an increased risk of pancreatic cancer (PaC) of similar magnitude, suggesting a common mechanism. The recent finding that PaC incidence increases linearly with increasing fasting glucose levels supports a central role for hyperglycaemia, which is known to cause carbonyl stress and advanced glycation end-product (AGE) accumulation through increased glycolytic activity and non-enzymatic reactions. This study investigated the impact of hyperglycaemia on invasive tumour development and the underlying mechanisms involved. Methods Pdx1-Cre;LSL-KrasG12D/+ mice were interbred with mitosis luciferase reporter mice, rendered diabetic with streptozotocin and treated or not with carnosinol (FL-926-16), a selective scavenger of reactive carbonyl species (RCS) and, as such, an inhibitor of AGE formation. Mice were monitored for tumour development by in vivo bioluminescence imaging. At the end of the study, pancreatic tissue was collected for histology/immunohistochemistry and molecular analyses. Mechanistic studies were performed in pancreatic ductal adenocarcinoma cell lines challenged with high glucose, glycolysis- and glycoxidation-derived RCS, their protein adducts AGEs and sera from diabetic patients. Results Cumulative incidence of invasive PaC at 22 weeks of age was 75% in untreated diabetic vs 25% in FL-926-16-gtreated diabetic and 8.3% in non-diabetic mice. FL-926-16 treatment suppressed systemic and pancreatic carbonyl stress, extracellular signal-regulated kinases (ERK) 1/2 activation, and nuclear translocation of Yes-associated protein (YAP) in pancreas. In vitro, RCS scavenging and AGE elimination completely inhibited cell proliferation stimulated by high glucose, and YAP proved essential in mediating the effects of both glucose-derived RCS and their protein adducts AGEs. However, RCS and AGEs induced YAP activity through distinct pathways, causing reduction of Large Tumour Suppressor Kinase 1 and activation of the Epidermal Growth Factor Receptor/ERK signalling pathway, respectively. Conclusions An RCS scavenger and AGE inhibitor prevented the accelerating effect of diabetes on PainINs progression to invasive PaC, showing that hyperglycaemia promotes PaC mainly through increased carbonyl stress. In vitro experiments demonstrated that both circulating RCS/AGEs and tumour cell-derived carbonyl stress generated by excess glucose metabolism induce proliferation by YAP activation, hence providing a molecular mechanism underlying the link between diabetes and PaC (and cancer in general).
Collapse
Affiliation(s)
- Stefano Menini
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Via di Grottarossa, 1035-1039 -, 00189, Rome, Italy
| | - Carla Iacobini
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Via di Grottarossa, 1035-1039 -, 00189, Rome, Italy
| | - Luisa de Latouliere
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Via di Grottarossa, 1035-1039 -, 00189, Rome, Italy.,SAFU-unit, Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Isabella Manni
- SAFU-unit, Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Martina Vitale
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Via di Grottarossa, 1035-1039 -, 00189, Rome, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Via di Grottarossa, 1035-1039 -, 00189, Rome, Italy.,Pathology Unit, University "La Sapienza", Sant'Andrea Hospital, Rome, Italy
| | - Carlo Pesce
- DINOGMI, University of Genoa Medical School, Genoa, Italy
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Francesco Novelli
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Giulia Piaggio
- SAFU-unit, Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Giuseppe Pugliese
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Via di Grottarossa, 1035-1039 -, 00189, Rome, Italy.
| |
Collapse
|
15
|
Lee M, Sun J, Han M, Cho Y, Lee JY, Nam CM, Kang ES. Nationwide Trends in Pancreatitis and Pancreatic Cancer Risk Among Patients With Newly Diagnosed Type 2 Diabetes Receiving Dipeptidyl Peptidase 4 Inhibitors. Diabetes Care 2019; 42:2057-2064. [PMID: 31431452 DOI: 10.2337/dc18-2195] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 07/29/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Dipeptidyl peptidase 4 inhibitors (DPP-4i) are useful incretin-based antidiabetes drugs. However, there is a concern that DPP-4i may adversely impact the exocrine pancreas, owing to their pleiotropic effects. In this study, we investigated whether DPP-4i are associated with pancreatitis and pancreatic cancer using a nationwide population-based cohort study. RESEARCH DESIGN AND METHODS We included patients newly diagnosed with type 2 diabetes who were treated with antidiabetes drugs (n = 33,208) from 2007 to 2013. The data were obtained from the Korean National Health Insurance Service-Health Screening Cohort database (n = 514,866). Risk was estimated using a Cox proportional hazards model with time-dependent covariates. A 6-month lag time was used to account for a possible latency time. The risk across various time segments since the first prescription of DPP-4i was also analyzed. RESULTS Out of 33,208 subjects, 10,218 were new users of DPP-4i and 22,990 were new users of other antidiabetes drugs. DPP-4i significantly increased the risks of pancreatitis (adjusted hazard ratio [aHR] 1.24, 95% CI 1.01-1.52; P = 0.037) and pancreatic cancer (aHR 1.81, 95% CI 1.16-2.82; P = 0.009) with a 6-month drug use lag period. The risk of pancreatitis and pancreatic cancer was generally consistent in the first 12 months and 1 year after the initial prescription without showing an increasing trend according to exposure duration. CONCLUSIONS DPP-4i use is associated with increased risks of pancreatitis and pancreatic cancer in patients with newly diagnosed type 2 diabetes. However, the absence of increasing trend according to exposure duration suggests the chances of reverse causality, and long-term pancreatic safety of DPP-4i has to be further investigated.
Collapse
Affiliation(s)
- Minyoung Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jiyu Sun
- Biostatistics and Computing, Graduate School, Yonsei University, Seoul, Republic of Korea
| | - Minkyung Han
- Department of Public Health, Graduate School, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yongin Cho
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji-Yeon Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chung Mo Nam
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Seok Kang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea .,Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
16
|
Li Y, Bian X, Wei S, He M, Yang Y. The relationship between pancreatic cancer and type 2 diabetes: cause and consequence. Cancer Manag Res 2019; 11:8257-8268. [PMID: 31571983 PMCID: PMC6750859 DOI: 10.2147/cmar.s211972] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/23/2019] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PC) is a devastating and lethal malignant disease and it is well known that there is a complex bidirectional relationship between PC and type 2 diabetes mellitus (T2DM). In order to more deeply summarize the relationship between them, this article summarizes the epidemiological data on the relationship between PC and T2DM in the past 5 years, and further explains the mechanism of interaction between them. Meanwhile, it also summed up the effects of drug therapy for T2DM on PC and the impact of T2DM on surgical resection of PC. Epidemiological studies clearly indicate that the risk of PC is increased in patients with T2DM. But increasing epidemiological data points out that PC also acts as a cause of T2DM and new-onset T2DM is sign and consequence of PC. Insulin resistance, hyperinsulinemia, hyperglycemia, and chronic inflammation are the mechanisms of T2DM-Associated PC. Metformin decreases the risk of PC, while insulin therapy increases the risk of PC. Besides, studies have shown that T2DM decreases the survival in patients with PC resection.
Collapse
Affiliation(s)
- Yan Li
- Department of Gerontology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Xiaohui Bian
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Shuyi Wei
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Meizhi He
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Yuelian Yang
- Department of Gerontology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
17
|
Dietary oleic acid is inversely associated with pancreatic cancer - Data from food diaries in a cohort study. Pancreatology 2018; 18:655-660. [PMID: 30031691 DOI: 10.1016/j.pan.2018.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 07/06/2018] [Accepted: 07/08/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Dietary oleic acid may prevent pancreatic ductal adenocarcinoma (PDA) by reducing hyperinsulinaemia which can otherwise promote DNA damage and tumour growth. Results from previous epidemiological studies investigating oleic acid are inconsistent. This study aims to clarify the relationship between dietary oleic acid intake and the risk of developing PDA using nutritional information from food diaries plus published serum biomarker data from HbA1c. METHODS 23,658 participants, aged 40-74 years, were recruited into EPIC-Norfolk and completed 7-day food diaries which recorded; foods, brands and portion sizes to calculate nutrient intakes. Serum HbA1c was measured at recruitment in 11,147 participants (48.7% of cohort). Hazard ratios (HRs) for quintiles of dietary oleic acid intake and serum HbA1c were estimated using Cox regression. Additional analyses were made according to whether body mass index (BMI) was greater or less than 25 kg/m2 as this influences hyperinsulinaemia. RESULTS 88 participants (55% women) developed PDA after a mean follow-up of 8.4 years (SD = 3.9) (mean age at diagnosis = 72.6 years, SD = 8.8). A decreased risk of PDA was associated with increased dietary oleic acid intake (highest vs lowest quintile, HR = 0.29, 95% CI = 0.10-0.81, P trend across quintiles = 0.011), with statistical significance maintained when BMI>25 kg/m2 but not if BMI<25 kg/m2. An elevated serum HbA1c was associated with increased risk of disease (highest vs lowest quintiles, HR = 6.32, 95% CI = 1.38-28.89, P for trend = 0.004). CONCLUSIONS The data supports a protective role of oleic acid against development of PDA in those with higher BMIs possibly through influencing hyperinsulinaemia. Oleic acid intake should be accurately measured in future aetiological studies.
Collapse
|
18
|
Huang BZ, Le Marchand L, Haiman CA, Monroe KR, Wilkens LR, Zhang ZF, Setiawan VW. Atopic allergic conditions and pancreatic cancer risk: Results from the Multiethnic Cohort Study. Int J Cancer 2018; 142:2019-2027. [PMID: 29314003 DOI: 10.1002/ijc.31241] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/09/2017] [Accepted: 12/15/2017] [Indexed: 01/14/2023]
Abstract
Previous case-control studies have suggested that atopic allergic conditions (AACs) are inversely associated with pancreatic cancer, but this relationship has not been supported in many prospective settings. In this study, we investigated the influence of AACs (asthma, hay fever, or allergy) and the treatment of these conditions on pancreatic cancer risk among participants of the Multiethnic Cohort Study (MEC). AACs and antihistamine use were assessed via a baseline questionnaire when participants joined the MEC in 1993-1996. Risk ratios (RRs) and 95% confidence intervals (CIs) for pancreatic cancer incidence by AACs and antihistamines were calculated using Cox regression, adjusting for age, sex, ethnicity, education, smoking status, family history of pancreatic cancer, body mass index, diabetes, and alcohol intake. We further evaluated associations among subgroups defined by age, sex, ethnicity, follow-up time, and known pancreatic cancer risk factors. During an average 16-year follow-up, 1,455 incident cases of pancreatic cancer were identified among 187,226 white, African American, Latino, Japanese American, and Native Hawaiian men and women. AACs (RR 1.00, 95% CI 0.88-1.12) and antihistamines (RR 0.92, 95% CI 0.78-1.07) were not clearly associated with pancreatic cancer incidence. While these associations were also null for most subgroups, we did observe protective associations of AACs (RR 0.74, 95% CI 0.56-0.98) and antihistamines (RR 0.66, 95% CI 0.45-0.96) among the oldest participants (70+). Our results, in agreement with past prospective studies, suggest that AACs are not associated with pancreatic cancer in general, but the observed protective associations among the oldest age group may warrant future investigation.
Collapse
Affiliation(s)
- Brian Z Huang
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA.,Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA.,Norris Comprehensive Cancer Center, Los Angeles, CA
| | - Kristine R Monroe
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Lynne R Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI
| | - Zuo-Feng Zhang
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA
| | - Veronica Wendy Setiawan
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA.,Norris Comprehensive Cancer Center, Los Angeles, CA
| |
Collapse
|
19
|
Abstract
OBJECTIVES Presentation of pancreatic adenocarcinoma (PC) as acute pancreatitis (AP), association of chronic pancreatitis (CP) with PC, and role of inflammation in PC carcinogenesis are well recognized. We hypothesized that inflammatory changes associated with remote history of AP (≥2 years before PC diagnosis) would result in earlier age of PC diagnosis. METHODS We evaluated PC patients prospectively enrolled in the Pancreatic Adenocarcinoma Gene Environment Risk (PAGER) study at the University of Pittsburgh for history of pancreatitis and reviewed relevant medical records and imaging studies. Univariate and multivariable linear regression analyses evaluated the relationship between PC and remote history of AP. RESULTS Among 790 patients with histologically confirmed PC, 114 (14.4%) had a history of pancreatitis (AP within 2 years of PC diagnosis in 69 [8.7%], remote history of AP in 28 [3.5%], CP in 4 [0.5%], and unknown duration of pancreatitis in 13 [1.6%]). After controlling for age, sex, body mass index, smoking, alcohol history, and diabetic status at diagnosis, patients with a remote history of AP were diagnosed on average 4.7 years earlier with PC when compared with PC patients without history of AP (P < 0.035). CONCLUSIONS Remote history of AP may accelerate carcinogenesis in PC.
Collapse
|
20
|
Vora J, Ray K, Kosiborod M, Poulter NR, Rajagopalan S, Leiter LA. Cancer risks of anti-hyperglycemic drugs for type 2 diabetes treatment - a clinical appraisal. J Diabetes Complications 2017; 31:1451-1457. [PMID: 28655490 DOI: 10.1016/j.jdiacomp.2017.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/07/2017] [Accepted: 06/07/2017] [Indexed: 12/18/2022]
Abstract
AIM A clinical appraisal of existing scientific literature sought to assess the need for long-term prospective epidemiological studies to investigate an increased cancer risk of anti-hyperglycemic medication in type 2 diabetes. METHOD A focus statement was formulated as: "With a higher risk of cancers in patients with type 2 diabetes, all anti-hyperglycemic drugs should undergo long-term, prospective epidemiological studies for cancer risks." Field surveys were sent to practicing physicians and endocrinologists to identify the currently prevalent level of acceptance of this statement. Subsequently, a meeting with a six-member panel of key opinion leaders was held to discuss published evidence in support and against the statement. This publication reviews the publications and discussion points brought forth in this meeting and their effect on statement acceptance by the panel. RESULTS Whereas the majority of field survey responders primarily agreed with the statement, panel members were divided in their statement support. This division remained intact after review of the literature. CONCLUSIONS While there was evidence that type 2 diabetes is associated with an increased risk of cancer, existing studies seemed insufficient to definitively demonstrate a link between cancer risk and use of specific anti-hyperglycemic therapies.
Collapse
Affiliation(s)
- Jiten Vora
- Department of Diabetes and Endocrinology, Royal Liverpool University Hospitals, Prescot Street, Liverpool, L7 8XP, UK.
| | - Kausik Ray
- Department of Primary Care and Public Health, School of Public Health, Imperial College London, St. Dunstan's Road, London, W6 8RP, UK.
| | - Mikhail Kosiborod
- Saint Luke's Mid America Heart Institute, University of Missouri-Kansas City, 4401 Wornall Road, Kansas City, MO 64111, USA.
| | - Neil R Poulter
- International Centre for Circulatory Health, Imperial College London, London, W2 1PG, UK.
| | - Sanjay Rajagopalan
- University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201, USA.
| | - Lawrence A Leiter
- Division of Endocrinology & Metabolism, Li Ka Shing Knowledge Institute and Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, 61 Queen St. East #6121, Toronto, ON, M5C 2T2, Canada.
| |
Collapse
|
21
|
Shindo K, Yu J, Suenaga M, Fesharakizadeh S, Tamura K, Almario JAN, Brant A, Borges M, Siddiqui A, Datta L, Wolfgang CL, Hruban RH, Klein AP, Goggins M. Lack of association between the pancreatitis risk allele CEL-HYB and pancreatic cancer. Oncotarget 2017; 8:50824-50831. [PMID: 28881607 PMCID: PMC5584208 DOI: 10.18632/oncotarget.15137] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 01/01/2017] [Indexed: 12/20/2022] Open
Abstract
CEL-HYB is a hybrid allele that arose from a crossover between the 3’ end of the Carboxyl ester lipase (CEL) gene and the nearby CEL pseudogene (CELP) and was recently identified as a risk factor for chronic pancreatitis. Since chronic pancreatitis is a risk factor for the development of pancreatic cancer, we compared the prevalence of the CEL-HYB allele in patients with pancreatic ductal adenocarcinoma to spousal controls and disease controls. The CEL-HYB allele was detected using Sanger and next generation sequencing. There was no significant difference in the prevalence of the CEL-HYB allele between cases with pancreatic ductal adenocarcinoma compared to controls; 2.6% (22/850) vs. 1.8% (18/976) (p=0.35). CEL-HYB carriers were not more likely to report a history of pancreatitis. Patients with pancreatic cancer are not more likely than controls to be carriers of the CEL-HYB allele.
Collapse
Affiliation(s)
- Koji Shindo
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jun Yu
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Masaya Suenaga
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shahriar Fesharakizadeh
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Koji Tamura
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jose Alejandro Navarro Almario
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aaron Brant
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael Borges
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Abdulrehman Siddiqui
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lisa Datta
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christopher L Wolfgang
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ralph H Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alison Patricia Klein
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Epidemiology, Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Michael Goggins
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
22
|
Gomez-Rubio P, Zock JP, Rava M, Marquez M, Sharp L, Hidalgo M, Carrato A, Ilzarbe L, Michalski C, Molero X, Farré A, Perea J, Greenhalf W, O'Rorke M, Tardón A, Gress T, Barberà V, Crnogorac-Jurcevic T, Domínguez-Muñoz E, Muñoz-Bellvís L, Alvarez-Urturi C, Balcells J, Barneo L, Costello E, Guillén-Ponce C, Kleeff J, Kong B, Lawlor R, Löhr M, Mora J, Murray L, O'Driscoll D, Peláez P, Poves I, Scarpa A, Real FX, Malats N. Reduced risk of pancreatic cancer associated with asthma and nasal allergies. Gut 2017; 66:314-322. [PMID: 26628509 DOI: 10.1136/gutjnl-2015-310442] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/26/2015] [Accepted: 10/28/2015] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Studies indicate an inverse association between ductal adenocarcinoma of the pancreas (PDAC) and nasal allergies. However, controversial findings are reported for the association with asthma. Understanding PDAC risk factors will help us to implement appropriate strategies to prevent, treat and diagnose this cancer. This study assessed and characterised the association between PDAC and asthma and corroborated existing reports regarding the association between allergies and PDAC risk. DESIGN Information about asthma and allergies was collated from 1297 PDAC cases and 1024 controls included in the PanGenEU case-control study. Associations between PDAC and atopic diseases were studied using multilevel logistic regression analysis. Meta-analyses of association studies on these diseases and PDAC risk were performed applying random-effects model. RESULTS Asthma was associated with lower risk of PDAC (OR 0.64, 95% CI 0.47 to 0.88), particularly long-standing asthma (>=17 years, OR 0.39, 95% CI 0.24 to 0.65). Meta-analysis of 10 case-control studies sustained our results (metaOR 0.73, 95% CI 0.59 to 0.89). Nasal allergies and related symptoms were associated with lower risk of PDAC (OR 0.66, 95% CI 0.52 to 0.83 and OR 0.59, 95% CI 0.46 to 0.77, respectively). These results were supported by a meta-analysis of nasal allergy studies (metaOR 0.6, 95% CI 0.5 to 0.72). Skin allergies were not associated with PDAC risk. CONCLUSIONS This study shows a consistent inverse association between PDAC and asthma and nasal allergies, supporting the notion that atopic diseases are associated with reduced cancer risk. These results point to the involvement of immune and/or inflammatory factors that may either foster or restrain pancreas carcinogenesis warranting further research to understand the molecular mechanisms driving this association.
Collapse
Affiliation(s)
- Paulina Gomez-Rubio
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Jan-Paul Zock
- Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
| | - Marta Rava
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Mirari Marquez
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Linda Sharp
- National Cancer Registry Ireland, Cork, Ireland, and Institute of Health & Society, Newcastle University, UK
| | | | - Alfredo Carrato
- Department of Oncology, Hospital Ramón y Cajal, Madrid, Spain
| | - Lucas Ilzarbe
- Hospital del Mar-Parc de Salut Mar, Barcelona, Spain
| | | | - Xavier Molero
- Exocrine Pancreas Research Unit, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Antoni Farré
- Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - José Perea
- Department of Surgery, 12 de Octubre University Hospital, Madrid, Spain
| | | | - Michael O'Rorke
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Adonina Tardón
- Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | - Thomas Gress
- Department of Gastroenterology, University Hospital Giessen and Marburg, Marburg, Germany
| | - Victor Barberà
- Laboratorio de Genética Molecular, Hospital General Universitario de Elche, Elche, Spain
| | | | | | - Luís Muñoz-Bellvís
- Cirugía General y del Aparato Digestivo, Hospital Universitario de Salamanca, Salamanca, Spain
| | | | - Joaquim Balcells
- Exocrine Pancreas Research Unit, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Luis Barneo
- Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain
| | | | | | - Jörg Kleeff
- Technical University of Munich, Munich, Germany
| | - Bo Kong
- Technical University of Munich, Munich, Germany
| | - Rita Lawlor
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Matthias Löhr
- Gastrocentrum, Karolinska Institutet, Stockholm, Sweden
| | - Josefina Mora
- Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Lim Murray
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Damian O'Driscoll
- National Cancer Registry Ireland, Cork, Ireland, and Institute of Health & Society, Newcastle University, UK
| | - Pablo Peláez
- Department of Surgery, 12 de Octubre University Hospital, Madrid, Spain
| | - Ignasi Poves
- Hospital del Mar-Parc de Salut Mar, Barcelona, Spain
| | - Aldo Scarpa
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| |
Collapse
|
23
|
Sarnecka AK, Zagozda M, Durlik M. An Overview of Genetic Changes and Risk of Pancreatic Ductal Adenocarcinoma. J Cancer 2016; 7:2045-2051. [PMID: 27877219 PMCID: PMC5118667 DOI: 10.7150/jca.15323] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 06/02/2016] [Indexed: 12/17/2022] Open
Abstract
The pancreatic carcinoma is a leading cause of death in cancer carriers worldwide. The early diagnostic is difficult due to late stage during diagnosis, lack of characteristic symptoms and also multifactor basis. In cancer development take part both, environmental and genetic factors, alone or in conjunction with each other. The nonspecific biomarkers of cancers are a reason for the search for more accurate factors which allow for fast and personalized diagnostics. Some of cancers have identified molecular (metabolic, biochemical or genetic) markers but in most cases the only clue is patient`s interview and abnormal levels of organ functions markers. Possible genetic basis of cancer suggests to widen studies on connection between environmental factors with both, nuclear and mitochondrial, genes changes.
Collapse
Affiliation(s)
- Agnieszka K Sarnecka
- Department of Surgical Research & Transplantology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.; Central Clinical Hospital, Ministry of Internal Affairs, Warsaw, Poland
| | - Malgorzata Zagozda
- Department of Surgical Research & Transplantology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.; Central Clinical Hospital, Ministry of Internal Affairs, Warsaw, Poland
| | - Marek Durlik
- Department of Surgical Research & Transplantology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.; Central Clinical Hospital, Ministry of Internal Affairs, Warsaw, Poland
| |
Collapse
|
24
|
Midha S, Chawla S, Garg PK. Modifiable and non-modifiable risk factors for pancreatic cancer: A review. Cancer Lett 2016; 381:269-277. [PMID: 27461582 DOI: 10.1016/j.canlet.2016.07.022] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/10/2016] [Accepted: 07/19/2016] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma is associated with a poor prognosis and a high case-fatality rate. The reasons for poor prognosis are low rates of curative resection due to local infiltration and distant metastasis. To increase survival rates of patients with pancreatic cancer, early detection through surveillance and screening is important. However, screening could only be cost-effective in high-risk populations. Identification of significant risk factors therefore assumes significance. Risk factors could be non-modifiable or modifiable. Non-modifiable risk factors include increasing age, familial cancer syndromes, Afro-American race, hereditary and other forms of chronic pancreatitis, diabetes, and non-O blood group. Important modifiable risk factors include smoking, obesity, dietary factors such as non-vegetarian diet, and toxins. Preventive strategies at the population level and an effective screening program targeted at high-risk people may help in prevention and early detection of pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Shallu Midha
- Department of Gastrointestinal Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Saurabh Chawla
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Pramod Kumar Garg
- Department of Gastroenterology, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
25
|
Abstract
Gallstones, particularly cholesterol gallstones, are common in Western populations and may cause symptoms such as biliary colic or complications such as acute cholecystitis or gallstone pancreatitis. Recent studies have allowed for a better understanding of the risk of symptoms or complications in patients with gallstones. In addition, newer data suggest an association of gallstones with overall mortality, cardiovascular disease, gastrointestinal cancers, and non-alcoholic fatty liver disease. Knowledge of appropriate indications and timing of cholecystectomy, particularly for mild biliary pancreatitis, has gradually accumulated. Lastly, there are exciting possibilities for novel agents to treat or prevent cholesterol stone disease. This review covers new advances in our understanding of the natural history, clinical associations, and management of gallstone disease.
Collapse
Affiliation(s)
- Evan Tiderington
- Division of Gastroenterology, University of Washington, Seattle, WA, USA
| | - Sum P Lee
- Division of Gastroenterology, University of Washington, Seattle, WA, USA
| | - Cynthia W Ko
- Division of Gastroenterology, University of Washington, Seattle, WA, USA
| |
Collapse
|
26
|
Camara SN, Yin T, Yang M, Li X, Gong Q, Zhou J, Zhao G, Yang ZY, Aroun T, Kuete M, Ramdany S, Camara AK, Diallo AT, Feng Z, Ning X, Xiong JX, Tao J, Qin Q, Zhou W, Cui J, Huang M, Guo Y, Gou SM, Wang B, Liu T, Olivier OET, Conde T, Cisse M, Magassouba AS, Ballah S, Keita NLM, Souare IS, Toure A, Traore S, Balde AK, Keita N, Camara ND, Emmanuel D, Wu HS, Wang CY. High risk factors of pancreatic carcinoma. JOURNAL OF HUAZHONG UNIVERSITY OF SCIENCE AND TECHNOLOGY. MEDICAL SCIENCES = HUA ZHONG KE JI DA XUE XUE BAO. YI XUE YING DE WEN BAN = HUAZHONG KEJI DAXUE XUEBAO. YIXUE YINGDEWEN BAN 2016; 36:295-304. [PMID: 27376795 DOI: 10.1007/s11596-016-1583-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 05/15/2016] [Indexed: 12/20/2022]
Abstract
Over the past decades, cancer has become one of the toughest challenges for health professionals. The epidemiologists are increasingly directing their research efforts on various malignant tumor worldwide. Of note, incidence of cancers is on the rise more quickly in developed countries. Indeed, great endeavors have to be made in the control of the life-threatening disease. As we know it, pancreatic cancer (PC) is a malignant disease with the worst prognosis. While little is known about the etiology of the PC and measures to prevent the condition, so far, a number of risk factors have been identified. Genetic factors, pre-malignant lesions, predisposing diseases and exogenous factors have been found to be linked to PC. Genetic susceptibility was observed in 10% of PC cases, including inherited PC syndromes and familial PC. However, in the remaining 90%, their PC might be caused by genetic factors in combination with environmental factors. Nonetheless, the exact mechanism of the two kinds of factors, endogenous and exogenous, working together to cause PC remains poorly understood. The fact that most pancreatic neoplasms are diagnosed at an incurable stage of the disease highlights the need to identify risk factors and to understand their contribution to carcinogenesis. This article reviews the high risk factors contributing to the development of PC, to provide information for clinicians and epidemiologists.
Collapse
Affiliation(s)
- Soriba Naby Camara
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Yin
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ming Yang
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiang Li
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiong Gong
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Zhou
- Department of Breast and Thyroid Surgery, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gang Zhao
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhi-Yong Yang
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tajoo Aroun
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Martin Kuete
- Department of Planning Family and Reproductive Institute, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sonam Ramdany
- Department of General Medicine, Sir Seewoosagur Ramgoolam National Hospital of Pamplemousses, Mauritius, 21017, Mauritius
| | | | - Aissatou Taran Diallo
- Department of General Surgery, National Hospital of Ignace Deen, Conakry, 1147, Guinea
| | - Zhen Feng
- Department of Gastroenterology and Hepatology, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xin Ning
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiong-Xin Xiong
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Tao
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qi Qin
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Zhou
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Cui
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Min Huang
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yao Guo
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shan-Miao Gou
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bo Wang
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Liu
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ohoya Etsaka Terence Olivier
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tenin Conde
- Department of Thoracic Surgery, National Hospital of Donka, Conakry, Guinea
| | - Mohamed Cisse
- Department of Dermatology, National Hospital of Donka, Conakry, Guinea
| | | | - Sneha Ballah
- Department of Internal Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Naby Laye Moussa Keita
- Department of Biochemistry, University Gamal Abdel Nasser of Conakry, Conakry, 1147, Guinea
| | - Ibrahima Sory Souare
- Department of Neurosurgery, Friendship Hospital Sino-Guinea of Kipe, Conakry, Guinea
| | - Aboubacar Toure
- Department of General Surgery, National Hospital of Ignace Deen, Conakry, 1147, Guinea
| | - Sadamoudou Traore
- Department of Medical Imaging, Good Shepherd Medical Center, The University of Texas, Longview, 75601, USA
| | | | - Namory Keita
- Department of Gynecology and Obstetrics, National Hospital of Donka, Conakry, Guinea
| | - Naby Daouda Camara
- Department of General Surgery, National Hospital of Ignace Deen, Conakry, 1147, Guinea
| | - Dusabe Emmanuel
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - He-Shui Wu
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun-You Wang
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW A relevant number of patients with pancreatic disorders suffer from secondary diabetes. Recent data have shed light on the link between pancreatic damage and subsequent impairments in glucose homeostasis. Furthermore, epidemiological studies provided insights into the relationship between diabetes and the risk of pancreatic carcinoma or pancreatitis. Pancreaticogenic diabetes requires a tailored therapeutic approach taking into account the individual properties of the available glucose-lowering drugs. RECENT FINDINGS We review the available literature concerning diabetes in patients with acute or chronic pancreatitis or pancreatic carcinoma. The relationship between the pancreatic damage and alterations in insulin and glucose homeostasis is summarized as well as the effect of diabetes mellitus on the risk of pancreatic cancer and pancreatitis. Caveats in the treatment of pancreaticogenic diabetes with currently available drugs are being discussed. SUMMARY Patients with pancreatic diseases should be screened for diabetes by means of an oral glucose tolerance test. There is a close inverse relationship between pancreatic β-cell loss and postchallenge hyperglycemia. The risk of hypoglycemia may be increased in patients with pancreaticogenic diabetes. Newly diagnosed diabetes may be a harbinger of pancreatic cancer. There is increasing evidence suggesting an increased risk for (pancreatic) cancer and pancreatitis in patients with diabetes mellitus. Further studies on the ideal glucose-lowering treatment of patients with pancreaticogenic diabetes will be required.
Collapse
|
28
|
Song S, Wang B, Zhang X, Hao L, Hu X, Li Z, Sun S. Long-Term Diabetes Mellitus Is Associated with an Increased Risk of Pancreatic Cancer: A Meta-Analysis. PLoS One 2015. [PMID: 26222906 PMCID: PMC4519136 DOI: 10.1371/journal.pone.0134321] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Previous studies have shown a bidirectional relationship between diabetes and pancreatic cancer (PC). In particular, new-onset diabetes might be induced by PC, and people with long-term diabetes might be at increased risk for the development of PC. The purpose of our study was to examine whether long-term diabetes represented an independent risk factor for PC development. Methodology A literature search was performed by searching electronic databases for studies published before July 1, 2014, and relative risks (RRs) and corresponding 95% confidence intervals (CIs) were calculated. Data pertaining to diabetes were recorded at both individual and study levels, with RRs calculated separately to analyze the relationship between the duration of diabetes and the development of PC. Results Forty-four studies were included in this meta-analysis, including 18 studies with a case-control design, 5 with a nested case-control design and 21 with a cohort design. The overall summary estimate for the relationship between the population with a duration of diabetes ≥2 years and PC was 1.64 (1.52-1.78). The pooled RR (95% CI) of PC for the population with a duration of diabetes ≥5 years was 1.58 (1.42-1.75). For the population with a duration of diabetes ≥10 years, the RR (95% CI) of PC was 1.50 (1.28-1.75). Conclusions Our study suggests that long-term diabetes mellitus is associated with an increased risk of PC. However, the level of risk is negatively correlated with increasing diabetes mellitus duration.
Collapse
Affiliation(s)
- Shanshan Song
- Department of Pancreas and Breast Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Baosheng Wang
- Department of Pancreas and Breast Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xin Zhang
- Department of Pancreas and Breast Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liliang Hao
- Department of Pancreas and Breast Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xianliang Hu
- Department of Pancreas and Breast Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhongxiang Li
- Department of Pancreas and Breast Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shaolong Sun
- Department of Pancreas and Breast Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- * E-mail:
| |
Collapse
|
29
|
Salvatore T, Marfella R, Rizzo MR, Sasso FC. Pancreatic cancer and diabetes: A two-way relationship in the perspective of diabetologist. Int J Surg 2015; 21 Suppl 1:S72-7. [PMID: 26123386 DOI: 10.1016/j.ijsu.2015.06.063] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 03/25/2015] [Accepted: 04/10/2015] [Indexed: 12/31/2022]
Abstract
Diabetes is a risk factor for pancreatic cancer as roughly half of all patients with pancreatic cancer are found to have diabetes at time of diagnosis. Moreover, an around 2-fold risk of pancreatic malignancy in diabetic patients has even be recently resulted from two meta-analysis. Actually, there is a bidirectional association between the two entities that implies a complex and reverse causality. In fact, while the risk for pancreatic cancer is modestly but significantly increased in patients with long-standing diabetes, recent-onset diabetes appears to be very frequently associated with pancreatic malignancy. Therefore, diabetes could serve as an excellent clue for early detection of pancreatic cancer. Moreover, recent epidemiological findings support the hypothesis that chronic exposure to hyperglycemia, higher insulin concentrations, and insulin resistance may be responsible for the enhanced risk of developing pancreatic cancer. Epidemiological data suggest that the type of anti-diabetic therapy may affect the risk of developing pancreatic cancer. In particular, metformin has been shown to reduce the risk of pancreatic cancer, as well as several other malignancies. On the other hand, some hypoglycemic agents could determine an increase of pancreatic cancer risk. These last findings were not confirmed. Finally, pancreatic cancer necessitates of a multidisciplinary management, primarily including surgeons and oncologists. In this context, the diabetologist plays an important role, given that his actions may influence the prevention and early diagnosis of pancreatic cancer, the perioperative complications associated to glycemic derangement, as well as the proper treatment of postpancreactomy diabetes.
Collapse
Affiliation(s)
- Teresa Salvatore
- Depart. of Internal and Experimental Medicine "Magrassi - Lanzara", Second University of Naples, Italy.
| | - Raffaele Marfella
- Depart. of Medicine, Surgery, Neurology, Metabolism and Geriatrics, Second University of Naples, Italy.
| | - Maria Rosaria Rizzo
- Depart. of Medicine, Surgery, Neurology, Metabolism and Geriatrics, Second University of Naples, Italy.
| | - Ferdinando Carlo Sasso
- Depart. of Internal and Experimental Medicine "Magrassi - Lanzara", Second University of Naples, Italy.
| |
Collapse
|
30
|
Tong GX, Geng QQ, Chai J, Cheng J, Chen PL, Liang H, Shen XR, Wang DB. Association between pancreatitis and subsequent risk of pancreatic cancer: a systematic review of epidemiological studies. Asian Pac J Cancer Prev 2015; 15:5029-34. [PMID: 24998582 DOI: 10.7314/apjcp.2014.15.12.5029] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This study aimed to summarize published epidemiological evidence for the relationship between pancreatitis and subsequent risk of pancreatic cancer (PC). We searched Medline and Embase for epidemiological studies published by February 5th, 2014 examining the risk of PC in pancreatitis patients using highly inclusive algorithms. Information about first author, year of publication, country of study, recruitment period, type of pancreatitis, study design, sample size, source of controls and attained age of subjects were extracted by two researchers and Stata 11.0 was used to perform the statistical analyses and examine publication bias. Odds ratios (ORs) with 95% confidence intervals (CIs) were calculated with the random effects model. A total of 17 articles documenting 3 cohort and 14 case-control studies containing 14,667 PC cases and 17,587 pancreatitis cases were included in this study. The pooled OR between pancreatitis and PC risk was 7.05 (95%CI: 6.42-7.75). However, the pooled ORs of case-control and cohort studies were 4.62 (95%CI: 4.08-5.22) and 16.3 (95%CI: 14.3-18.6) respectively. The risk of PC was the highest in patients with chronic pancreatitis (pooled OR=10.35; 95%CI: 9.13-11.75), followed by unspecified type of pancreatitis (pooled OR=6.41; 95%CI: 4.93-8.34), both acute and chronic pancreatitis (pooled OR=6.13; 95%CI: 5.00-7.52), and acute pancreatitis (pooled OR=2.12; 95%CI: 1.59-2.83). The pooled OR of PC in pancreatitis cases diagnosed within 1 year was the highest (pooled OR=23.3; 95%CI: 14.0-38.9); and the risk in subjects diagnosed with pancreatitis for no less than 2, 5 and 10 years were 3.03 (95%CI: 2.41-3.81), 2.82 (95%CI: 2.12-3.76) and 2.25 (95%CI: 1.59-3.19) respectively. Pancreatitis, especially chronic pancreatitis, was associated with a significantly increased risk of PC; and the risk decreased with increasing duration since diagnosis of pancreatitis.
Collapse
Affiliation(s)
- Gui-Xian Tong
- Center for Health Management, School of Health Services Management, Anhui Medical University, Hefei, China E-mail :
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Bosetti C, Rosato V, Li D, Silverman D, Petersen GM, Bracci PM, Neale RE, Muscat J, Anderson K, Gallinger S, Olson SH, Miller AB, Bas Bueno-de-Mesquita H, Scelo G, Janout V, Holcatova I, Lagiou P, Serraino D, Lucenteforte E, Fabianova E, Baghurst PA, Zatonski W, Foretova L, Fontham E, Bamlet WR, Holly EA, Negri E, Hassan M, Prizment A, Cotterchio M, Cleary S, Kurtz RC, Maisonneuve P, Trichopoulos D, Polesel J, Duell EJ, Boffetta P, La Vecchia C, Ghadirian P. Diabetes, antidiabetic medications, and pancreatic cancer risk: an analysis from the International Pancreatic Cancer Case-Control Consortium. Ann Oncol 2014; 25:2065-2072. [PMID: 25057164 PMCID: PMC4176453 DOI: 10.1093/annonc/mdu276] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 06/30/2014] [Accepted: 07/01/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus has been associated with an excess risk of pancreatic cancer, but the magnitude of the risk and the time-risk relationship are unclear, and there is limited information on the role of antidiabetic medications. PATIENTS AND METHODS We analyzed individual-level data from 15 case-control studies within the Pancreatic Cancer Case-Control Consortium, including 8305 cases and 13 987 controls. Pooled odds ratios (ORs) were estimated from multiple logistic regression models, adjusted for relevant covariates. RESULTS Overall, 1155 (15%) cases and 1087 (8%) controls reported a diagnosis of diabetes 2 or more years before cancer diagnosis (or interview, for controls), corresponding to an OR of 1.90 (95% confidence interval, CI, 1.72-2.09). Consistent risk estimates were observed across strata of selected covariates, including body mass index and tobacco smoking. Pancreatic cancer risk decreased with duration of diabetes, but a significant excess risk was still evident 20 or more years after diabetes diagnosis (OR 1.30, 95% CI 1.03-1.63). Among diabetics, long duration of oral antidiabetic use was associated with a decreased pancreatic cancer risk (OR 0.31, 95% CI 0.14-0.69, for ≥15 years). Conversely, insulin use was associated with a pancreatic cancer risk in the short term (OR 5.60, 95% CI 3.75-8.35, for <5 years), but not for longer duration of use (OR 0.95, 95% CI 0.53-1.70, for ≥15 years). CONCLUSION This study provides the most definitive quantification to date of an excess risk of pancreatic cancer among diabetics. It also shows that a 30% excess risk persists for more than two decades after diabetes diagnosis, thus supporting a causal role of diabetes in pancreatic cancer. Oral antidiabetics may decrease the risk of pancreatic cancer, whereas insulin showed an inconsistent duration-risk relationship.
Collapse
Affiliation(s)
- C Bosetti
- Department of Epidemiology, IRCCS - Istituto di Ricerche Farmacologiche 'Mario Negri', Milan, Italy.
| | - V Rosato
- Department of Epidemiology, IRCCS - Istituto di Ricerche Farmacologiche 'Mario Negri', Milan, Italy
| | - D Li
- M.D. Anderson Cancer Center, University of Texas, Houston
| | - D Silverman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda
| | - G M Petersen
- Department of Health Sciences Research, Medicine and Medical Genetics, Mayo Clinic, Rochester
| | - P M Bracci
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, USA
| | - R E Neale
- Queensland Institute of Medical Research, Brisbane, Australia
| | - J Muscat
- Department of Public Health Sciences, Penn State University, Penn State
| | - K Anderson
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, USA
| | - S Gallinger
- University Health Network, Department of Surgery, University of Toronto, Toronto, Canada
| | - S H Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - A B Miller
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - H Bas Bueno-de-Mesquita
- National Institute for Public Health and the Environment (RIVM), Bilthoven; Department of Gastroenterology and Hepatology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands; Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - G Scelo
- International Agency for Research on Cancer (IARC), Lyon, France
| | - V Janout
- Department of Preventive Medicine, Faculty of Medicine, Palacky University, Olomouc
| | - I Holcatova
- Institute of Hygiene and Epidemiology, 1st Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - P Lagiou
- Department of Epidemiology, Harvard School of Public Health, Boston, USA; Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, University of Athens, Athens, Greece
| | - D Serraino
- Unit of Epidemiology and Biostatistics, CRO Aviano National Cancer Institute, IRCCS, Aviano
| | - E Lucenteforte
- Department of Preclinical and Clinical Pharmacology Mario Aiazzi Mancini, Università degli Studi di Firenze, Florence, Italy
| | - E Fabianova
- Regional Authority of Public Health in Banská Bystrica, Banská Bystrica, Slovakia
| | - P A Baghurst
- Public Health, Women's and Children's Hospital, Adelaide, SA, Australia
| | - W Zatonski
- Cancer Center and Institute of Oncology, Warsaw, Poland
| | - L Foretova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Institute and MF MU, Brno, Czech Republic
| | - E Fontham
- Louisiana State University School of Public Health, New Orleans, USA
| | - W R Bamlet
- Department of Health Sciences Research, Medicine and Medical Genetics, Mayo Clinic, Rochester
| | - E A Holly
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, USA
| | - E Negri
- Department of Epidemiology, IRCCS - Istituto di Ricerche Farmacologiche 'Mario Negri', Milan, Italy
| | - M Hassan
- M.D. Anderson Cancer Center, University of Texas, Houston
| | - A Prizment
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, USA
| | - M Cotterchio
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada; Cancer Care Ontario, Toronto, Canada
| | - S Cleary
- University Health Network, Department of Surgery, University of Toronto, Toronto, Canada
| | - R C Kurtz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - P Maisonneuve
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| | - D Trichopoulos
- Department of Epidemiology, Harvard School of Public Health, Boston, USA
| | - J Polesel
- Unit of Epidemiology and Biostatistics, CRO Aviano National Cancer Institute, IRCCS, Aviano
| | - E J Duell
- Unit of Nutrition, Environment and Cancer, Catalan Institute of Oncology (ICO-IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - P Boffetta
- The Tisch Cancer Institute and Institute for Translational Epidemiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - C La Vecchia
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
32
|
Cholelithiasis and risk of pancreatic cancer: systematic review and meta-analysis of 21 observational studies. Cancer Causes Control 2014; 25:1543-51. [DOI: 10.1007/s10552-014-0458-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 08/06/2014] [Indexed: 12/31/2022]
|
33
|
Starup-Linde J, Karlstad O, Eriksen SA, Vestergaard P, Bronsveld HK, de Vries F, Andersen M, Auvinen A, Haukka J, Hjellvik V, Bazelier MT, Boer AD, Furu K, De Bruin ML. CARING (CAncer Risk and INsulin analoGues): the association of diabetes mellitus and cancer risk with focus on possible determinants - a systematic review and a meta-analysis. Curr Drug Saf 2014; 8:296-332. [PMID: 24215312 PMCID: PMC5421136 DOI: 10.2174/15748863113086660071] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 10/27/2013] [Accepted: 10/30/2013] [Indexed: 12/11/2022]
Abstract
Background: Patients suffering from diabetes mellitus (DM) may experience an increased risk of cancer; however, it is not certain whether this effect is due to diabetes per se. Objective: To examine the association between DM and cancers by a systematic review and meta-analysis according to the PRISMA guidelines. Data Sources: The systematic literature search includes Medline at PubMed, Embase, Cinahl, Bibliotek.dk, Cochrane library, Web of Science and SveMed+ with the search terms: “Diabetes mellitus”, “Neoplasms”, and “Risk of cancer”. Study Eligibility Criteria: The included studies compared the risk of cancer in diabetic patients versus non-diabetic patients. All types of observational study designs were included. Results: Diabetes patients were at a substantially increased risk of liver (RR=2.1), and pancreas (RR=2.2) cancer. Modestly elevated significant risks were also found for ovary (RR=1.2), breast (RR=1.1), cervix (RR=1.3), endometrial (RR=1.4), several digestive tract (RR=1.1-1.5), kidney (RR=1.4), and bladder cancer (RR=1.1). The findings were similar for men and women, and unrelated to study design. Meta-regression analyses showed limited effect modification of body mass index, and possible effect modification of age, gender, with some influence of study characteristics (population source, cancer- and diabetes ascertainment). Limitations: Publication bias seemed to be present. Only published data were used in the analyses. Conclusions: The systematic review and meta-analysis confirm the previous results of increased cancer risk in diabetes and extend this to additional cancer sites. Physicians in contact with patients with diabetes should be aware that diabetes patients are at an increased risk of cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Marie L De Bruin
- Department of Endocrinology and Internal Medicine (MEA), Aarhus University Hospital, Tage Hansens Gade 2, 8000 Aarhus C, Denmark.
| |
Collapse
|
34
|
Musolino C, Allegra A, Minciullo PL, Gangemi S. Allergy and risk of hematologic malignancies: associations and mechanisms. Leuk Res 2014; 38:1137-44. [PMID: 25171954 DOI: 10.1016/j.leukres.2014.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/02/2014] [Accepted: 08/04/2014] [Indexed: 12/12/2022]
Abstract
Increasing evidence indicates that a dysregulated immune system, as the one found in allergic disorders, can affect survival of tumor cells. A possible association between allergies and risk of hematologic malignancies has been examined in several epidemiological studies; however, results were not always consistent. The aim of this review is to report the preclinical and clinical data, which support a correlation between allergy and hematologic neoplasms. Immune system modulation could represent a powerful tool in the prevention and treatment of hematologic malignancies.
Collapse
Affiliation(s)
- C Musolino
- Division of Hematology, Department of General Surgery and Oncology, University of Messina, Messina, Italy
| | - A Allegra
- Division of Hematology, Department of General Surgery and Oncology, University of Messina, Messina, Italy.
| | - P L Minciullo
- School and Division of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University Hospital "G. Martino", Messina, Italy
| | - S Gangemi
- School and Division of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University Hospital "G. Martino", Messina, Italy; Institute of Clinical Physiology, IFC CNR, Messina Unit, Messina, Italy
| |
Collapse
|
35
|
He XD, Wu Q, Liu W, Hong T, Li JJ, Miao RY, Zhao HT. Association of metabolic syndromes and risk factors with ampullary tumors development: A case-control study in China. World J Gastroenterol 2014; 20:9541-9548. [PMID: 25071350 PMCID: PMC4110587 DOI: 10.3748/wjg.v20.i28.9541] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 02/22/2014] [Accepted: 05/19/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To evaluate the risk factors for ampullary adenoma and ampullary cancer.
METHODS: This case-control study included ampullary tumor patients referred to Peking Union Medical College Hospital. Controls were randomly selected from an existing database of healthy individuals at the Health Screening Center of the same hospital. Data on metabolic syndromes, medical conditions, and family history were collected by retrospective review of the patients’ records and health examination reports, or by interview.
RESULTS: A total of 181 patients and 905 age- and sex-matched controls were enrolled. We found that a history of diabetes, cholecystolithiasis, low-density lipoprotein, and apolipoprotein A were significantly related to ampullary adenomas. Diabetes, cholecystolithiasis, chronic pancreatitis, total cholesterol, high-density lipoprotein, and apolipoprotein A were also significantly related to ampullary cancer.
CONCLUSION: Some metabolic syndrome components and medical conditions are potential risk factors for the development of ampullary tumors. Cholelithiasis, diabetes, and apolipoprotein A may contribute to the malignant transformation of benign ampullary adenomas into ampullary cancer.
Collapse
|
36
|
Nogueira L, Freedman ND, Engels EA, Warren JL, Castro F, Koshiol J. Gallstones, cholecystectomy, and risk of digestive system cancers. Am J Epidemiol 2014; 179:731-9. [PMID: 24470530 DOI: 10.1093/aje/kwt322] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gallstones and cholecystectomy may be related to digestive system cancer through inflammation, altered bile flux, and changes in metabolic hormone levels. Although gallstones are recognized causes of gallbladder cancer, associations with other cancers of the digestive system are poorly established. We used the Surveillance, Epidemiology, and End Results (SEER)-Medicare linked database (1992-2005), which includes 17 cancer registries that cover approximately 26% of the US population, to identify first primary cancers (n = 236,850) occurring in persons aged ≥66 years and 100,000 cancer-free population-based controls frequency-matched by calendar year, age, and gender. Odds ratios and 95% confidence intervals were calculated using logistic regression analysis, adjusting for the matching factors. Gallstones and cholecystectomy were associated with increased risk of noncardia gastric cancer (odds ratio (OR) = 1.21 (95% confidence interval (CI): 1.11, 1.32) and OR = 1.26 (95% CI: 1.13, 1.40), respectively), small-intestine carcinoid (OR = 1.27 (95% CI: 1.01, 1.60) and OR = 1.78 (95% CI: 1.41, 2.25)), liver cancer (OR = 2.35 (95% CI: 2.18, 2.54) and OR = 1.26 (95% CI: 1.12, 1.41)), and pancreatic cancer (OR = 1.24 (95% CI: 1.16, 1.31) and OR = 1.23 (95% CI: 1.15, 1.33)). Colorectal cancer risk associated with gallstones and cholecystectomy decreased with increasing distance from the common bile duct (P-trend < 0.001). Hence, gallstones and cholecystectomy are associated with the risk of cancers occurring throughout the digestive tract.
Collapse
|
37
|
Preziosi G, Oben JA, Fusai G. Obesity and pancreatic cancer. Surg Oncol 2014; 23:61-71. [PMID: 24746917 DOI: 10.1016/j.suronc.2014.02.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 02/21/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Pancreatic cancer is an invariably fatal malignancy. Cigarette smoking and diabetes are established risk factors, but over the last two decades studies have shown that excess adiposity is an additional independent risk factor with 30-50% of cases thought to be attributed to nutritional factors. The aim of this narrative review is to analyze all the epidemiological evidence on the topic and possible pathophysiology. METHODS We searched PubMed, Embase, Cochrane Library and Medline, and all available evidence was included. We firstly analyze meta- and pooled analysis. Then we discuss individual studies to identify sources of discrepancies between studies and attempt to delineate pathophysiology. RESULTS It is estimated that obese individuals have a relative risk (RR) ranging between 1.19 and 1.47, when compared with those of normal weight, regardless of diabetes or smoking status. No significant differences were found between gender. CONCLUSION There is a measurable increased risk of developing pancreatic cancer in obese individuals, and excess adiposity is related to the condition with a "dose-response" curve. Hyperinsulinemia and possibly hyperestrogenism secondary to a metabolic syndrome, and independently from diabetes status, appear to be the key elements of the pathogenesis in pancreatic cancer secondary to excess body fat. Increased efforts should therefore be made in tackling the epidemic levels of obesity in the Western world countries.
Collapse
Affiliation(s)
- Giuseppe Preziosi
- Hepato-Pancreatico-Biliary Surgery and Liver Transplant Unit, Royal Free Hospital, London, United Kingdom.
| | - Jude A Oben
- Centre for Liver and Digestive Health, University College London, Royal Free Hospital, London, United Kingdom
| | - Giuseppe Fusai
- Hepato-Pancreatico-Biliary Surgery and Liver Transplant Unit, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
38
|
Batabyal P, Vander Hoorn S, Christophi C, Nikfarjam M. Association of diabetes mellitus and pancreatic adenocarcinoma: a meta-analysis of 88 studies. Ann Surg Oncol 2014; 21:2453-62. [PMID: 24609291 DOI: 10.1245/s10434-014-3625-6] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is often diagnosed at an advanced, incurable stage. Previous epidemiological data suggests that diabetes mellitus (DM) is a risk factor for PDAC, which may be important in early detection. However, the strength of this association needs to be determined, taking into account a number of recently published studies. METHODS A systematic review of the association between DM and PDAC was undertaken by searching electronic databases and journal references from 1973 to 2013. Summary estimates were obtained separately for case-control and cohort studies by means of a 'random effects' approach. Data pertaining to the DM was recorded and plotted at both an individual and study level, with the relative risks (RR) pooled separately to determine the relationship of DM duration and PDAC. RESULTS A total of 88 independent studies, including 50 cohort and 39 case-control studies were examined. The overall summary-combined RR was 1.97 (95 % CI 1.78-2.18) with marked heterogeneity that could not be clearly attributed to any subgroup analyses. The risk of PDAC was greatest early after the diagnosis of DM but remained elevated long after the diagnosis. The individual-level RR ranged from 6.69 at less than 1 year to 1.36 at 10 years. CONCLUSION The results demonstrate a strong association between PDAC and recently diagnosed DM, which may be attributed to a paraneoplastic effect. However, the presence of diabetes also remains a modest risk factor for the development of PDAC long-term. Selective screening of patients with new-onset DM for PDAC needs to be considered.
Collapse
Affiliation(s)
- Pikli Batabyal
- Department of Surgery, University of Melbourne, Austin Health, LTB 8, 145 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
| | | | | | | |
Collapse
|
39
|
Oikonomopoulou K, Brinc D, Hadjisavvas A, Christofi G, Kyriacou K, Diamandis EP. The bifacial role of helminths in cancer: Involvement of immune and non-immune mechanisms. Crit Rev Clin Lab Sci 2014; 51:138-48. [DOI: 10.3109/10408363.2014.886180] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
40
|
Cotterchio M, Lowcock E, Hudson TJ, Greenwood C, Gallinger S. Association between allergies and risk of pancreatic cancer. Cancer Epidemiol Biomarkers Prev 2014; 23:469-80. [PMID: 24554712 DOI: 10.1158/1055-9965.epi-13-0965] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Less than 10% of pancreatic cancer cases survive 5 years, yet its etiology is not well understood. Studies suggest allergies are associated with reduced pancreatic cancer risk. Our study collected additional information on allergies (including skin prick test results and differentiation of allergic/nonallergic asthma), and is the first to assess possible confounding by allergy medications. METHODS A population-based case-control study was designed to comprehensively assess the association between allergy and pancreatic cancer risk. Pancreas cancer cases were diagnosed during 2011 to 2012, and identified through the Ontario Cancer Registry (345 cases). Population-based controls were identified using random digit dialing and age/sex frequency matched to cases (1,285 controls). Questionnaires collected lifetime allergy history (type of allergy, age at onset, skin prick testing results), allergy medications, and established pancreas cancer risk factors. Logistic regression was used to estimate odd ratios and test potential confounders, including allergy medications. RESULTS Hay fever was associated with a significant reduction in pancreatic cancer risk [AOR = 0.68; 95% confidence intervals (CI), 0.52-0.89], and reduction was greatest for those whose skin prick test was positive for hay fever allergens. No particular patterns were observed as regards age at onset and duration of allergy. Positive dust/mold allergy skin prick test and animal allergies were associated with a statistically significant reduced pancreatic cancer risk; AOR = 0.49; 95% CI, 0.31-0.78 and AOR = 0.68; 95% CI, 0.46-0.99, respectively. Asthma was not associated with pancreatic cancer risk. CONCLUSIONS/IMPACT These findings support the growing body of evidence that suggests certain allergies are associated with reduced pancreatic cancer risk.
Collapse
Affiliation(s)
- Michelle Cotterchio
- Authors' Affiliations: Prevention and Cancer Control, Cancer Care Ontario; Dalla Lana School of Public Health, University of Toronto; Departments of Medical Biophysics and Molecular Genetics, University of Toronto; Ontario Institute for Cancer Research; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital; and Division of General Surgery, Toronto General Hospital, Toronto, Ontario; and Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | | | | | | | | |
Collapse
|
41
|
Ahn J, Lee JK. [New-onset diabetes as an early sign of pancreatic cancer]. THE KOREAN JOURNAL OF GASTROENTEROLOGY 2013; 62:263-6. [PMID: 24262590 DOI: 10.4166/kjg.2013.62.5.263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
While long-standing diabetes is a risk factor of pancreatic cancer, new-onset diabetes could be a consequence of underlying pancreatic malignancy. About 30% to 50% of pancreatic cancer patients have new-onset diabetes. Because diabetes develops in preclinical or early stages of pancreatic cancer, it could serve as an excellent clue for early detection of pancreatic cancer. Insulin resistance associated with hyperglycemia and hyperinsulinemia by diabetogenic factors secreted from cancer cells have been suggested to be a possible mechanism of pancreatic cancer-induced diabetes. It is difficult to differentiate pancreatic cancer-induced diabetes from the more common type 2 diabetes. Although several clinical features and potential biomarkers have been investigated, optimal strategies and modalities to screen pancreatic cancer among the new-onset diabetes have not yet been fully determined.
Collapse
Affiliation(s)
- Jemma Ahn
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | |
Collapse
|
42
|
Olson SH, Hsu M, Satagopan JM, Maisonneuve P, Silverman DT, Lucenteforte E, Anderson KE, Borgida A, Bracci PM, Bueno-de-Mesquita HB, Cotterchio M, Dai Q, Duell EJ, Fontham EH, Gallinger S, Holly EA, Ji BT, Kurtz RC, La Vecchia C, Lowenfels AB, Luckett B, Ludwig E, Petersen GM, Polesel J, Seminara D, Strayer L, Talamini R. Allergies and risk of pancreatic cancer: a pooled analysis from the Pancreatic Cancer Case-Control Consortium. Am J Epidemiol 2013; 178:691-700. [PMID: 23820785 PMCID: PMC3755648 DOI: 10.1093/aje/kwt052] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 03/01/2013] [Indexed: 12/23/2022] Open
Abstract
In order to quantify the risk of pancreatic cancer associated with history of any allergy and specific allergies, to investigate differences in the association with risk according to age, gender, smoking status, or body mass index, and to study the influence of age at onset, we pooled data from 10 case-control studies. In total, there were 3,567 cases and 9,145 controls. Study-specific odds ratios and 95% confidence intervals were calculated by using unconditional logistic regression adjusted for age, gender, smoking status, and body mass index. Between-study heterogeneity was assessed by using the Cochran Q statistic. Study-specific odds ratios were pooled by using a random-effects model. The odds ratio for any allergy was 0.79 (95% confidence interval (CI): 0.62, 1.00) with heterogeneity among studies (P < 0.001). Heterogeneity was attributable to one study; with that study excluded, the pooled odds ratio was 0.73 (95% CI: 0.64, 0.84) (Pheterogeneity = 0.23). Hay fever (odds ratio = 0.74, 95% CI: 0.56, 0.96) and allergy to animals (odds ratio = 0.62, 95% CI: 0.41, 0.94) were related to lower risk, while there was no statistically significant association with other allergies or asthma. There were no major differences among subgroups defined by age, gender, smoking status, or body mass index. Older age at onset of allergies was slightly more protective than earlier age.
Collapse
Affiliation(s)
- Sara H Olson
- Department of Epidemiology and Biostatistics, 307 East 63rd Street, New York, NY 10065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Pezzilli R, Pagano N. Is diabetes mellitus a risk factor for pancreatic cancer? World J Gastroenterol 2013; 19:4861-4866. [PMID: 23946590 PMCID: PMC3740415 DOI: 10.3748/wjg.v19.i30.4861] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 04/21/2013] [Accepted: 05/08/2013] [Indexed: 02/06/2023] Open
Abstract
The relationship between diabetes mellitus and the risk of pancreatic cancer has been a matter of study for a long period of time. The importance of this topic is due to two main causes: the possible use of recent onset diabetes as a marker of the disease and, in particular, as a specific marker of pancreatic cancer, and the selection of a population at risk for pancreatic cancer. Thus, we decided to make an in-depth study of this topic; thus, we carried out an extensive literature search in order to re-assess the current knowledge on this topic. Even if diabetes is found a decade before the appearance of pancreatic cancer as reported in meta-analytic studies, we cannot select those patients already having non detectable pancreatic cancer, at least with the imaging and biological techniques available today. We believe that more studies are necessary in order to definitively identify diabetes mellitus as a risk factor for pancreatic cancer taking into consideration that approximately 10 years are needed to diagnose symptomatic pancreatic cancer. At present, the answer to the as to whether diabetes and pancreatic cancer comes first similar to the adage of the chicken and the egg is that diabetes is the egg.
Collapse
|
44
|
Abstract
Up to 85% of patients with pancreatic cancer have diabetes or hyperglycaemia, which frequently manifests as early as 2-3 years before a diagnosis of pancreatic cancer. Conversely, patients with new-onset diabetes have a 5-8-fold increased risk of being diagnosed with pancreatic cancer within 1-3 years of developing diabetes. Emerging evidence now indicates that pancreatic cancer causes diabetes. As in type 2 diabetes, β-cell dysfunction and peripheral insulin resistance are seen in pancreatic cancer-induced diabetes. However, unlike in patients with type 2 diabetes, glucose control worsens in patients with pancreatic cancer in the face of ongoing, often profound, weight loss. Diabetes and weight loss, which precede cachexia onset by several months, are paraneoplastic phenomena induced by pancreatic cancer. Although the pathogenesis of these pancreatic cancer-induced metabolic alterations is only beginning to be understood, these are likely mechanisms to promote the survival and growth of pancreatic cancer in a hostile and highly desmoplastic microenvironment. Interestingly, these metabolic changes could enable early diagnosis of pancreatic cancer, if they can be distinguished from the ones that occur in patients with type 2 diabetes. One such possible biomarker is adrenomedullin, which is a potential mediator of β-cell dysfunction in pancreatic cancer-induced diabetes.
Collapse
|
45
|
Lai HC, Tsai IJ, Chen PC, Muo CH, Chou JW, Peng CY, Lai SW, Sung FC, Lyu SY, Morisky DE. Gallstones, a cholecystectomy, chronic pancreatitis, and the risk of subsequent pancreatic cancer in diabetic patients: a population-based cohort study. J Gastroenterol 2013; 48:721-7. [PMID: 23053420 DOI: 10.1007/s00535-012-0674-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 08/21/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND The causal association between diabetes and pancreatic cancer remains unclear in Asian populations. This study examined whether gallstones, a cholecystectomy, chronic pancreatitis and the treatment of antidiabetic agents affect the risk of subsequent pancreatic cancer for patients with diabetes in a Taiwanese population. METHODS Using claims data from the universal health insurance program in Taiwan, 449,685 newly diagnosed diabetic cases among insured people from 2000 to 2003 were identified as the case group. The comparison group, matched for gender, age, and the index year of the diabetes cohort, consisted of 325,729 persons without diabetes. Pancreatic cancer incidence was measured in both groups until the end of 2008. Other risk factors associated with this cancer were also measured. RESULTS The incidence of pancreatic cancer in the diabetic cohort was 2-fold greater than that in the comparison group (1.46 vs. 0.71 per 10,000 person-years) with an adjusted hazard ratio (HR) of 1.75 [95 % confidence interval (CI) 1.45-2.10]. The risk slightly increased for diabetic patients with gallstones, cholecystitis, and a cholecystectomy (HR 1.92, 95% CI 1.18-3.11), but greatly increased for those with comorbidity of chronic pancreatitis (HR 22.9, 95% CI 12.6-41.4). Pancreatic cancer risk also increased significantly for those patients who used more insulin for treating diabetes (OR 2.20, 95% CI 1.40-3.45). CONCLUSION Our data suggest that the risk of pancreatic cancer is moderately increased in patients with diabetes, especially those using insulin therapy. The risk is greatly increased for diabetic patients with chronic pancreatitis.
Collapse
Affiliation(s)
- Hsueh-Chou Lai
- School of Chinese Medicine, China Medical University, Taichung, 404, Taiwan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW The association of diabetes and cancer has received increased attention as data have emerged to indicate that the type of diabetes treatment may influence the risk of cancer, and that the risk of cancer among diabetic individuals can be reduced by intervention. The association of diabetes and pancreatic cancer is particularly strong, but often misunderstood. Long-standing type 1 diabetes and type 2 diabetes increase the risk for this malignancy, but the cancer can also induce pancreatogenic, or type 3c, diabetes as well. RECENT FINDINGS This review covers the recent findings which help to clarify these relationships, and offers guidance for prevention, early detection, and treatment. Obesity and, separately, diabetes increase the risk of several common malignancies by about two-fold. This risk is reduced by successful treatments. Type 3c diabetes is more common than previously realized, and strategies to differentiate type 3c diabetes from type 2 diabetes, to identify those candidates who will benefit from screening studies, are discussed. SUMMARY The death rate because of pancreatic and other cancers can be reduced by an aggressive approach to reversing obesity and hyperinsulinemia, achieving good glycemic control in diabetic patients, and identifying at an early timepoint those patients with pancreatogenic diabetes.
Collapse
MESH Headings
- Blood Glucose/metabolism
- Carcinoma, Pancreatic Ductal/blood
- Carcinoma, Pancreatic Ductal/etiology
- Carcinoma, Pancreatic Ductal/pathology
- Cell Transformation, Neoplastic
- Diabetes Complications/blood
- Diabetes Complications/pathology
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/pathology
- Early Detection of Cancer
- Female
- Humans
- Hyperinsulinism/blood
- Hyperinsulinism/complications
- Hypoglycemic Agents/adverse effects
- Male
- Metformin/administration & dosage
- Metformin/adverse effects
- Obesity/blood
- Obesity/complications
- Obesity/pathology
- Pancreatic Neoplasms/blood
- Pancreatic Neoplasms/etiology
- Pancreatic Neoplasms/pathology
- Peptides/blood
- Risk Factors
Collapse
Affiliation(s)
- Dana K Andersen
- Clinical Studies Program, Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
47
|
Oikonomopoulou K, Brinc D, Kyriacou K, Diamandis EP. Infection and cancer: revaluation of the hygiene hypothesis. Clin Cancer Res 2013; 19:2834-41. [PMID: 23536438 DOI: 10.1158/1078-0432.ccr-12-3661] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Several studies have shown that persistent infections and inflammation can favor carcinogenesis. At the same time, certain types of pathogens and antitumor immune responses can decrease the risk of tumorigenesis or lead to cancer regression. Infectious agents and their products can orchestrate a wide range of host immune responses, through which they may positively or negatively modulate cancer development and/or progression. The factors that direct this dichotomous influence of infection-mediated immunity on carcinogenesis are not well understood. Even though not universal, several previous reports have investigated the inverse link of pathogen-induced "benign" inflammation to carcinogenesis and various other pathologies, ranging from autoimmune diseases to allergy and cancer. Several models and ideas are discussed in this review, including the impact of decreased exposure to pathogens, as well as the influence of pathogen load, the timing of infection, and the type of instigated immune response on carcinogenesis. These phenomena should guide future investigations into identifying novel targets within the microbial and host proteome, which will assist in the development of cancer therapeutics and vaccine remedies, analogous to earlier efforts based on helminthic components for the prevention and/or treatment of several pathologies.
Collapse
Affiliation(s)
- Katerina Oikonomopoulou
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
48
|
Luckett BG, Su LJ, Rood JC, Fontham ETH. Cadmium exposure and pancreatic cancer in south Louisiana. JOURNAL OF ENVIRONMENTAL AND PUBLIC HEALTH 2012; 2012:180186. [PMID: 23319964 PMCID: PMC3540786 DOI: 10.1155/2012/180186] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 11/04/2012] [Indexed: 01/21/2023]
Abstract
Cadmium has been hypothesized to be a pancreatic carcinogen. We test the hypothesis that cadmium exposure is a risk factor for pancreatic cancer with a population-based case-control study sampled from a population with persistently high rates of pancreatic cancer (south Louisiana). We tested potential dietary and nondietary sources of cadmium for their association with urinary cadmium concentrations which reflect long-term exposure to cadmium due to the accumulation of cadmium in the kidney cortex. Increasing urinary cadmium concentrations were significantly associated with an increasing risk of pancreatic cancer (2nd quartile OR = 3.34, 3rd = 5.58, 4th = 7.70; test for trend P ≤ 0.0001). Potential sources of cadmium exposure, as documented in the scientific literature, found to be statistically significantly associated with increased risk of pancreatic cancer included working as a plumber, pipefitter or welder (OR = 5.88) and high consumption levels of red meat (4th quartile OR = 6.18) and grains (4th quartile OR = 3.38). Current cigarette smoking, at least 80 pack years of smoking, occupational exposure to cadmium and paints, working in a shipyard, and high consumption of grains were found to be statistically significantly associated with increased concentrations of urinary cadmium. This study provides epidemiologic evidence that cadmium is a potential human pancreatic carcinogen.
Collapse
Affiliation(s)
- Brian G Luckett
- Epidemiology Program, School of Public Health, Louisiana State University Health Sciences Center, 2020 Gravier Street, 3rd Floor, New Orleans, LA 70112, USA.
| | | | | | | |
Collapse
|
49
|
Abstract
Epidemiological studies clearly indicate that the risk of pancreatic cancer (PC) is increased in diabetic patients, but most studies focus on overall diabetes or type 2 diabetes mellitus (T2DM), and there are few studies on the risks of type 1 and type 3c (secondary) diabetes. Possible mechanisms for increased cancer risk in diabetes include cellular proliferative effects of hyperglycemia, hyperinsulinemia, and abnormalities in insulin/IGF receptor pathways. Recently, insulin and insulin secretagogues have been observed to increase the PC risk, while metformin treatment reduces the cancer risk in diabetic subjects. In addition, anticancer drugs used to treat PC may either cause diabetes or worsen coexisting diabetes. T3cDM has emerged as a major subset of diabetes and may have the highest risk of pancreatic carcinoma especially in patients with chronic pancreatitis. T3cDM is also a consequence of PC in at least 30% of patients. Distinguishing T3cDM from the more prevalent T2DM among new-onset diabetic patients can be aided by an assessment of clinical features and confirmed by finding a deficiency in postprandial pancreatic polypeptide release. In conclusion, diabetes and PC have a complex relationship that requires more clinical attention. The risk of developing PC can be reduced by aggressive prevention and treatment of T2DM and obesity and the prompt diagnosis of T3cDM may allow detection of a tumor at a potentially curable stage.
Collapse
Affiliation(s)
- YunFeng Cui
- Department of Surgery, Johns Hopkins Bayview Medical Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
50
|
Turner MC. Epidemiology: allergy history, IgE, and cancer. Cancer Immunol Immunother 2012; 61:1493-510. [PMID: 22183126 PMCID: PMC11028502 DOI: 10.1007/s00262-011-1180-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 12/01/2011] [Indexed: 01/10/2023]
Abstract
Numerous epidemiological studies have investigated potential associations between allergy history and cancer risk with strong inverse associations reported in studies of pancreatic cancer, glioma, and childhood leukemia. Recently, there has been a rapid expansion of the epidemiological literature both of studies evaluating self-reported allergy history in relation to cancer risk and of studies evaluating biological indicators of allergy history and immune function including levels of immunoglobulin (Ig) E. However, there are several potential methodological limitations associated with prior studies, and further research is required to clarify associations observed. This paper summarizes the recent epidemiological literature examining associations between allergy history and cancer risk. From 2008, a total of 55 epidemiological studies were identified that examined some aspect of the association between allergy and cancer. Although the majority of studies examined self-reported allergy history in relation to cancer risk, there were also studies examining allergy diagnoses or discharges as captured in existing administrative databases, levels of IgE, polymorphisms of allergy, inflammatory- or allergy-related cytokine genes, and concentrations of immune regulatory proteins. The most frequently studied cancer sites included brain and lymphatic and hematopoietic cancers. Potential methodological sources of bias are discussed as well as recommendations for future work.
Collapse
Affiliation(s)
- Michelle C Turner
- McLaughlin Centre for Population Health Risk Assessment, Institute of Population Health, University of Ottawa, One Stewart Street, Room 313, Ottawa, ON K1N 6N5, Canada.
| |
Collapse
|