1
|
Song L, Chen Z, Li Y, Ran L, Liao D, Zhang Y, Wang G. Trend and forecast analysis of the changing disease burden of pancreatic cancer attributable to high fasting glucose in China, 1990-2021. Front Oncol 2024; 14:1471699. [PMID: 39493456 PMCID: PMC11527594 DOI: 10.3389/fonc.2024.1471699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024] Open
Abstract
Background Pancreatic cancer (PC) is a malignant tumour with poor prognosis and high mortality, and high fasting plasma glucose (HFPG) is considered to be one of its important risk factors. Methods PC disease burden data were obtained from the Global Burden of Disease Study 2021 (GBD 2021) database. Annual percent change (APC), average APC (AAPC), and 95% confidence interval (95% CI) were analysed using joinpoint linkpoint regression models to assess the trend of PC burden of disease between 1990 and 2021. An age-period-cohort model was used to estimate the independent effects of age, period, and cohort on PC burden, and data on PC mortality attributable to HFPG in China from 2022 to 2032 were analysed on the basis of a Bayesian age-period-cohort model projection. Results The number of Pc deaths due to HFPG continue to rise in China from 1990 to 2021, with age-standardised mortality (ASMR) and age-standardised disability-adjusted life-year rates with increasing AAPC values of 1.12% (95% CI, 0.73-1.52) and 1.00% (95% CI, 0.63-1.37), respectively. Throughout the study, we found that the overall level of PC disease burden was significantly higher in men than that in women. In age-period-cohort analyses, the age effect of PC showed an increasing and then decreasing trend, the period effect showed an overall increasing trend during the study period, and the cohort effect showed an overall slow decreasing trend. In addition, the BAPC model predicted that ASMR is expected to decline significantly in both men and women from 2022 to 2032. Conclusions It was found that PC attributable to HFPG was generally on the rise in China from 1990 to 2021 and has been on the decline in recent years, and projections suggest that the country's future PC disease burden will continue to show a downward trend. Age and period of birth are the main factors affecting the disease burden, especially in men and older age groups. Early prevention, regular screening, and research into the pathogenesis of PC have, therefore, become particularly important.
Collapse
Affiliation(s)
- Lichen Song
- School of Clinical Medicine, Dali University, Dali, Yunnan, China
| | - Ziyi Chen
- School of Clinical Medicine, Dali University, Dali, Yunnan, China
| | - Yongjie Li
- School of Clinical Medicine, Dali University, Dali, Yunnan, China
| | - Lirong Ran
- School of Clinical Medicine, Dali University, Dali, Yunnan, China
| | - Dongwei Liao
- School of Clinical Medicine, Dali University, Dali, Yunnan, China
| | - Yuanyuan Zhang
- Medicine Department, School of Clinical Medicine, Dali University, Dali, Yunnan, China
| | - Guangming Wang
- Center of Genetic Testing, The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| |
Collapse
|
2
|
Huang X, Li H, Zhao L, Xu L, Long H. Prediabetes increases the risk of pancreatic cancer: A meta-analysis of longitudinal observational studies. PLoS One 2024; 19:e0311911. [PMID: 39405289 PMCID: PMC11478827 DOI: 10.1371/journal.pone.0311911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Glycemic disorder is closely related to the risk of pancreatic cancer, but previous studies focused on the influence of diabetes. The aim of this meta-analysis was to investigate the influence of prediabetes, an intermediate state between normoglycemia and diabetes, on the risk of pancreatic cancer. METHODS Relevant longitudinal observational studies were identified through a search of Medline, Embase, and Web of Science databases. To minimize the influence of between-study heterogeneity, a randomized-effects model was used to pool the results. RESULTS Nine cohort studies including 26,444,624 subjects were available for the meta-analysis. Among them, 2,052,986 (7.8%) had prediabetes at baseline, and the participants were followed for a mean duration of 5.9 years. It was found that, compared to people with normoglycemia, those with prediabetes had a higher incidence of pancreatic cancer (risk ratio [RR]: 1.42, 95% confidence interval: 1.36 to 1.49, p<0.001) with no statistical heterogeneity (I2 = 0%). Sensitivity analysis performed by excluding one dataset at a time did not significantly change the results (RR: 1.38 to 1.45, p all <0.05). Subgroup analyses indicated that the association between prediabetes and increased risk of pancreatic cancer was not significantly impacted by study characteristics such as study design, location, age, and sex of participants, definition of prediabetes, duration of follow-up, or adjustment for alcohol intake (p for subgroup difference all >0.05). CONCLUSIONS Prediabetes may be associated with an increased risk of pancreatic cancer compared to normoglycemia.
Collapse
Affiliation(s)
- Xuefang Huang
- Department of Gastroenterology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Huan Li
- Department of Gastroenterology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Lisha Zhao
- Department of Gastroenterology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Lingli Xu
- Department of Gastroenterology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Hui Long
- Department of Gastroenterology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
3
|
Li MX, Lopez-Aguiar AG, Poultsides G, Rocha F, Weber S, Fields R, Idrees K, Cho C, Maithel SK, Zhang XF, Pawlik TM. Preoperative Glycosylated Hemoglobin A1C Impacts Long-Term Outcomes Following Curative-Intent Resection of Nonfunctional Gastroenteropancreatic Neuroendocrine Tumors. J Surg Oncol 2024. [PMID: 39387595 DOI: 10.1002/jso.27951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND To investigate the impact of preoperative glycosylated hemoglobin A1C (HbA1c) among patients following curative-intent resection of nonfunctional gastroentropancreatic neuroendocrine tumors (GEP-NETs). METHODS Patients who underwent curative-intent resection for GEP-NETs from 2000 to 2020 were identified from the US Neuroendocrine Tumor Study Group (US-NETSG). Preoperative blood HbA1c levels were defined as high HbA1c (≥ 6.5%) versus low HbA1c group (< 6.5%). Impact of HbA1c level on postoperative short-term and long-term overall (OS) were investigated. RESULTS A total of 130 patients with HbA1c < 6.5% and 60 patients with HbA1c ≥ 6.5% were included. Patients with HbA1c ≥ 6.5% had higher proportion of comorbidities, such as hypertension, obesity, anemia, and lower preoperative albumin levels versus patients with HbA1c < 6.5% (all p < 0.05). In addition, high level of preoperative HbA1c was associated with increased incidence of wound and infectious complications, as well as decreased long-term OS (median OS: high Hb1Ac 89.8 months vs. low Hb1Ac not reached, HR 3.487, p = 0.004) among patients with nonfunctional GEP-NETs, as well as among the subset of pancreatic NET patients (median OS: high Hb1Ac 74.3 months vs. low Hb1Ac not reached, p = 0.004), and patients with normal fasting blood glucose (< 140 mg/dL) (median OS: high Hb1Ac 75.4 months vs. low Hb1Ac not reached, p = 0.001). CONCLUSIONS Hb1Ac might have value as a screening tool to identify high-risk patients following surgical resection of nonfunctional GEP-NETs for consideration of more strict postoperative surveillance and treatment of elevated Hb1Ac level.
Collapse
Affiliation(s)
- Mu-Xing Li
- Department of Hepatobiliary Surgery, Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Alexandra G Lopez-Aguiar
- Department of Surgery, Division of Surgical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - George Poultsides
- Department of Surgery, Stanford University, Palo Alto, California, USA
| | - Flavio Rocha
- Department of Surgery, Virginia Mason Medical Center, Seattle, Washington
| | - Sharon Weber
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Ryan Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, Wisconsin
| | - Kamran Idrees
- Department of Surgery, Division of Surgical Oncology, Vanderbilt University, Nashville, Tennessee, USA
| | - Cliff Cho
- Department of Surgery, Division of Hepatopancreatobiliary and Advanced Gastrointestinal Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Shishir K Maithel
- Department of Surgery, Division of Surgical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Xu-Feng Zhang
- Department of Hepatobiliary Surgery, Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Timothy M Pawlik
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, USA
| |
Collapse
|
4
|
McDonnell D, Cheang AWE, Wilding S, Wild SH, Frampton AE, Byrne CD, Hamady ZZ. Elevated Glycated Haemoglobin (HbA1c) Is Associated with an Increased Risk of Pancreatic Ductal Adenocarcinoma: A UK Biobank Cohort Study. Cancers (Basel) 2023; 15:4078. [PMID: 37627106 PMCID: PMC10452109 DOI: 10.3390/cancers15164078] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND The role of dysglycaemia as a risk marker for Pancreatic Ductal Adenocarcinoma (PDAC) is uncertain. We investigated the relationship between glycated haemoglobin (HbA1c) and incident PDAC using a retrospective cohort study within the UK Biobank. METHODS A study involving 499,804 participants from the UK Biobank study was undertaken. Participants were stratified by diabetes mellitus (DM) status, and then by HbA1c values < 42 mmol/mol, 42-47 mmol/mol, or ≥48 mmol/mol. Cox proportional hazard models were used to describe the association between HbA1c category (with time-varying interactions) and incident PDAC. RESULTS PDAC occurred in 1157 participants during 11.6 (10.9-12.3) years follow up [(median (interquartile range)]. In subjects without known DM at baseline, 12 months after recruitment, the adjusted hazard ratios (aHR, 95% CI) for incident PDAC for HbA1c 42-47 mmol/mol compared to HbA1c < 42 mmol/mol (reference group) was 2.10 (1.31-3.37, p = 0.002); and was 8.55 (4.58-15.99, p < 0.001) for HbA1c ≥ 48 mmol/mol. The association between baseline HbA1c and incident PDAC attenuated with increasing duration of time of follow-up to PDAC diagnosis. CONCLUSIONS Dysglycaemia detected by elevated HbA1c is associated with an increased risk of PDAC. The strength of the association between elevated HbA1c and incident PDAC is inversely proportional to the time from detecting dysglycaemia but remains significant for at least 60 months following HbA1c testing.
Collapse
Affiliation(s)
- Declan McDonnell
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
- HPB Unit, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Adrian W. E. Cheang
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
| | - Sam Wilding
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
| | - Sarah H. Wild
- Usher Institute, University of Edinburgh, Edinburgh EH8 9YL, UK;
| | - Adam E. Frampton
- Section of Oncology, University of Surrey, Guildford GU2 7XH, UK;
- HPB Unit, Royal Surrey NHS Foundation Trust, Guildford GU2 7XX, UK
| | - Christopher D. Byrne
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
| | - Zaed Z. Hamady
- Human Development & Health, University of Southampton, University Hospital, Southampton SO16 6YD, UK; (A.W.E.C.); (S.W.); (C.D.B.); (Z.Z.H.)
- HPB Unit, University Hospital Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
5
|
Sahoo OS, Mitra R, Bhattacharjee A, Kar S, Mukherjee O. Is Diabetes Mellitus a Predisposing Factor for Helicobacter pylori Infections? Curr Diab Rep 2023; 23:195-205. [PMID: 37213058 DOI: 10.1007/s11892-023-01511-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/23/2023] [Indexed: 05/23/2023]
Abstract
PURPOSE OF REVIEW This review aims to analyse the consistency of reports suggesting the role of Diabetes Mellitus in the pathogenesis of Helicobacter pylori (H. pylori). RECENT FINDINGS There have been numerous controversies citing the prevalence of H. pylori infections in patients suffering from type 2 diabetes mellitus (T2DM). This review investigates the possible crosstalk between H. pylori infections and T2DM and also designs a meta-analysis to quantify the association. Subgroup analyses have also been conducted to deduce factors like geography and testing techniques, in playing a role in stratification analysis. Based on a scientific literature survey and meta-analysis of databases from 1996 to 2022, a trend towards more frequent H. pylori infections in patients with diabetes mellitus was observed. The highly diversified nature of H. pylori infections across age, gender, and geographical regions requires large interventional studies to evaluate its long-term association with diabetes mellitus. Further possible linkage of the prevalence of diabetes mellitus concomitant with that of H. pylori infected patients has also been delineated in the review.
Collapse
Affiliation(s)
- Om Saswat Sahoo
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, India, 713209
| | - Rhiti Mitra
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, India, 713209
| | - Arghyadeep Bhattacharjee
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, India, 713209
- Department of Microbiology, Kingston College of Science, Beruanpukuria, Barasat, West Bengal, India, 700129
| | - Samarjit Kar
- Department of Mathematics, National Institute of Technology Durgapur, Durgapur, West Bengal, India, 713209
| | - Oindrilla Mukherjee
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, India, 713209.
| |
Collapse
|
6
|
Zhao R, Han Z, Zhou H, Xue Y, Chen X, Cao X. Diagnostic and prognostic role of circRNAs in pancreatic cancer: a meta-analysis. Front Oncol 2023; 13:1174577. [PMID: 37361594 PMCID: PMC10285410 DOI: 10.3389/fonc.2023.1174577] [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: 03/03/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Background Circular RNAs (circRNAs) are types of endogenous noncoding RNAs produced by selective splicing that are expressed highly specifically in various organisms and tissues and have numerous clinical implications in the regulation of cancer development and progression. Since circRNA is resistant to digestion by ribonucleases and has a long half-life, there is increasing evidence that circRNA can be used as an ideal candidate biomarker for the early diagnosis and prognosis of tumors. In this study, we aimed to reveal the diagnostic and prognostic value of circRNA in human pancreatic cancer (PC). Methods A systematic search for publications from inception to 22 July 2022 was conducted on Embase, PubMed, Web of Science (WOS), and the Cochrane Library databases. Available studies that correlated circRNA expression in tissue or serum with the clinicopathological, diagnostic, and prognostic values of PC patients were enrolled. Odds ratios (ORs) and corresponding 95% confidence intervals (CIs) were used to evaluate clinical pathological characteristics. Area under the curve (AUC), sensitivity, and specificity were adopted to assess diagnostic value. Hazard ratios (HRs) were utilized to assess disease-free survival (DFS) and overall survival (OS). Results This meta-analysis enrolled 32 eligible studies, including six on diagnosis and 21 on prognosis, which accounted for 2,396 cases from 245 references. For clinical parameters, high expression of carcinogenic circRNA was significantly associated with degree of differentiation (OR = 1.85, 95% CI = 1.47-2.34), TNM stage (OR = 0.46, 95% CI = 0.35-0.62), lymph node metastasis (OR = 0.39, 95% CI = 0.32-0.48), and distant metastasis (OR = 0.26, 95% CI = 0.13-0.51). As for clinical diagnostic utility, circRNA could discriminate patients with pancreatic cancer from controls, with an AUC of 0.86 (95% CI: 0.82-0.88), a relatively high sensitivity of 84%, and a specificity of 80% in tissue. In terms of prognostic significance, carcinogenic circRNA was correlated with poor OS (HR = 2.00, 95% CI: 1.76-2.26) and DFS (HR = 1.96, 95% CI: 1.47-2.62). Conclusion In summary, this study demonstrated that circRNA may act as a significant diagnostic and prognostic biomarker for pancreatic cancer.
Collapse
Affiliation(s)
- Ruihua Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhuo Han
- Department of General Surgery, Tangdu Hospital, The Air Force Medical University, Xi’an, China
| | - Haiting Zhou
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yaru Xue
- Department of Pediatric Medicine, Northwest Women’s and Children’s Hospital, Xi’an, China
| | - Xiaobing Chen
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinguang Cao
- Department of Digestive Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Mikolaskova I, Crnogorac-Jurcevic T, Smolkova B, Hunakova L. Nutraceuticals as Supportive Therapeutic Agents in Diabetes and Pancreatic Ductal Adenocarcinoma: A Systematic Review. BIOLOGY 2023; 12:158. [PMID: 36829437 PMCID: PMC9953002 DOI: 10.3390/biology12020158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
The correlation between pancreatic ductal adenocarcinoma (PDAC) and diabetes-related mechanisms support the hypothesis that early therapeutic strategies targeting diabetes can contribute to PDAC risk reduction and treatment improvement. A systematic review was conducted, using PubMed, Embase and Cochrane Library databases, to evaluate the current evidence from clinical studies qualitatively examining the efficacy of four natural products: Curcumin-Curcuma longa L.; Thymoquinone-Nigella sativa L.; Genistein-Glycine max L.; Ginkgo biloba L.; and a low-carbohydrate ketogenic diet in type 2 diabetes (T2D) and PDAC treatment. A total of 28 clinical studies were included, showing strong evidence of inter-study heterogeneity. Used as a monotherapy or in combination with chemo-radiotherapy, the studied substances did not significantly improve the treatment response of PDAC patients. However, pronounced therapeutic efficacy was confirmed in T2D. The natural products and low-carbohydrate ketogenic diet, combined with the standard drugs, have the potential to improve T2D treatment and thus potentially reduce the risk of cancer development and improve multiple biological parameters in PDAC patients.
Collapse
Affiliation(s)
- Iveta Mikolaskova
- Institute of Immunology, Faculty of Medicine, Comenius University, Odborarske Namestie 14, 811 08 Bratislava, Slovakia
| | - Tatjana Crnogorac-Jurcevic
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University, Charterhouse Square, London EC1M 6BQ, UK
| | - Bozena Smolkova
- Biomedical Research Center, Slovak Academy of Sciences, Cancer Research Institute, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Luba Hunakova
- Institute of Immunology, Faculty of Medicine, Comenius University, Odborarske Namestie 14, 811 08 Bratislava, Slovakia
| |
Collapse
|
8
|
Identification of the Genetic Association Between Type-2-Diabetes and Pancreatic Cancer. Biochem Genet 2022; 61:1143-1162. [DOI: 10.1007/s10528-022-10308-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022]
|
9
|
Tong Y, Sun M, Chen L, Wang Y, Li Y, Li L, Zhang X, Cai Y, Qie J, Pang Y, Xu Z, Zhao J, Zhang X, Liu Y, Tian S, Qin Z, Feng J, Zhang F, Zhu J, Xu Y, Lou W, Ji Y, Zhao J, He F, Hou Y, Ding C. Proteogenomic insights into the biology and treatment of pancreatic ductal adenocarcinoma. J Hematol Oncol 2022; 15:168. [PMID: 36434634 PMCID: PMC9701038 DOI: 10.1186/s13045-022-01384-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/02/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with poor prognosis. Proteogenomic characterization and integrative proteomic analysis provide a functional context to annotate genomic abnormalities with prognostic value. METHODS We performed an integrated multi-omics analysis, including whole-exome sequencing, RNA-seq, proteomic, and phosphoproteomic analysis of 217 PDAC tumors with paired non-tumor adjacent tissues. In vivo functional experiments were performed to further illustrate the biological events related to PDAC tumorigenesis and progression. RESULTS A comprehensive proteogenomic landscape revealed that TP53 mutations upregulated the CDK4-mediated cell proliferation process and led to poor prognosis in younger patients. Integrative multi-omics analysis illustrated the proteomic and phosphoproteomic alteration led by genomic alterations such as KRAS mutations and ADAM9 amplification of PDAC tumorigenesis. Proteogenomic analysis combined with in vivo experiments revealed that the higher amplification frequency of ADAM9 (8p11.22) could drive PDAC metastasis, though downregulating adhesion junction and upregulating WNT signaling pathway. Proteome-based stratification of PDAC revealed three subtypes (S-I, S-II, and S-III) related to different clinical and molecular features. Immune clustering defined a metabolic tumor subset that harbored FH amplicons led to better prognosis. Functional experiments revealed the role of FH in altering tumor glycolysis and in impacting PDAC tumor microenvironments. Experiments utilizing both in vivo and in vitro assay proved that loss of HOGA1 promoted the tumor growth via activating LARP7-CDK1 pathway. CONCLUSIONS This proteogenomic dataset provided a valuable resource for researchers and clinicians seeking for better understanding and treatment of PDAC.
Collapse
Affiliation(s)
- Yexin Tong
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Mingjun Sun
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Lingli Chen
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Yunzhi Wang
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Yan Li
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Lingling Li
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Xuan Zhang
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Yumeng Cai
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Jingbo Qie
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Yanrui Pang
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Ziyan Xu
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Jiangyan Zhao
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Xiaolei Zhang
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Yang Liu
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Sha Tian
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Zhaoyu Qin
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Jinwen Feng
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Fan Zhang
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Jiajun Zhu
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Yifan Xu
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Wenhui Lou
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Yuan Ji
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Jianyuan Zhao
- grid.16821.3c0000 0004 0368 8293Institute for Development and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children’s Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China ,grid.207374.50000 0001 2189 3846Department of Anatomy and Neuroscience Research Institute, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001 China
| | - Fuchu He
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China ,grid.419611.a0000 0004 0457 9072State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing, 102206 China ,grid.506261.60000 0001 0706 7839Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing, 102206 China
| | - Yingyong Hou
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| | - Chen Ding
- grid.8547.e0000 0001 0125 2443Institute of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Department of Pathology, Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200433 China
| |
Collapse
|
10
|
Lin J, Yin M, Liu L, Gao J, Yu C, Liu X, Xu C, Zhu J. The Development of a Prediction Model Based on Random Survival Forest for the Postoperative Prognosis of Pancreatic Cancer: A SEER-Based Study. Cancers (Basel) 2022; 14:cancers14194667. [PMID: 36230593 PMCID: PMC9563591 DOI: 10.3390/cancers14194667] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/20/2022] Open
Abstract
Simple Summary Surgery is the main treatment to cure pancreatic cancer (PC). However, the 5-year survival rate of surgical resection is only 10–20%. The aim of our study was to develop a prediction model with the novel machine learning algorithm random survival forest (RSF) and to offer easy-to-use prediction tools, including risk stratification and individual prognosis. The study would benefit patients and physicians in postoperative management and facilitate personalized medicine. Abstract Accurate prediction for the prognosis of patients with pancreatic cancer (PC) is a emerge task nowadays. We aimed to develop survival models for postoperative PC patients, based on a novel algorithm, random survival forest (RSF), traditional Cox regression and neural networks (Deepsurv), using the Surveillance, Epidemiology, and End Results Program (SEER) database. A total of 3988 patients were included in this study. Eight clinicopathological features were selected using least absolute shrinkage and selection operator (LASSO) regression analysis and were utilized to develop the RSF model. The model was evaluated based on three dimensions: discrimination, calibration, and clinical benefit. It found that the RSF model predicted the cancer-specific survival (CSS) of the postoperative PC patients with a c-index of 0.723, which was higher than the models built by Cox regression (0.670) and Deepsurv (0.700). The Brier scores at 1, 3, and 5 years (0.188, 0.177, and 0.131) of the RSF model demonstrated the model’s favorable calibration and the decision curve analysis illustrated the model’s value of clinical implement. Moreover, the roles of the key variables were visualized in the Shapley Additive Explanations plotting. Lastly, the prediction model demonstrates value in risk stratification and individual prognosis. In this study, a high-performance prediction model for PC postoperative prognosis was developed, based on RSF The model presented significant strengths in the risk stratification and individual prognosis prediction.
Collapse
Affiliation(s)
- Jiaxi Lin
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
- Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
| | - Minyue Yin
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
- Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
| | - Lu Liu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
- Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
| | - Jingwen Gao
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
- Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
| | - Chenyan Yu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
- Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
| | - Xiaolin Liu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
- Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
| | - Chunfang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
- Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
| | - Jinzhou Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
- Suzhou Clinical Center of Digestive Diseases, Suzhou 215000, China
- Correspondence:
| |
Collapse
|
11
|
Campbell PT, Newton CC, Jacobs EJ, McCullough ML, Wang Y, Rees-Punia E, Guinter MA, Murphy N, Koshiol J, Dehal AN, Rohan T, Strickler H, Petrick J, Gunter M, Zhang X, McGlynn KA, Pollak M, Patel AV, Gapstur SM. Prospective associations of hemoglobin A 1c and c-peptide with risk of diabetes-related cancers in the Cancer Prevention Study-II Nutrition Cohort. CANCER RESEARCH COMMUNICATIONS 2022; 2:653-662. [PMID: 36712480 PMCID: PMC9881454 DOI: 10.1158/2767-9764.crc-22-0082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/05/2022] [Accepted: 06/21/2022] [Indexed: 02/02/2023]
Abstract
Self-reported type 2 diabetes mellitus (T2DM) is a risk factor for many cancers, suggesting its pathology relates to carcinogenesis. We conducted a case-cohort study to examine associations of hemoglobin A1c (HbA1c) and c-peptide with cancers associated with self-reported T2DM. This study was drawn from a prospective cohort of 32,383 women and men who provided blood specimens at baseline: c-peptide and HbA1c were assessed in 3,000 randomly selected participants who were cancer-free-at-baseline and an additional 2,281 participants who were cancer-free-at-baseline and subsequently diagnosed with incident colorectal, liver, pancreatic, female breast, endometrial, ovarian, bladder, or kidney cancers. Weighted-Cox regression models estimated hazards ratios (HRs) and 95% confidence intervals (CI), adjusted for covariates. C-peptide was associated with higher risk of liver cancer (per standard deviation (SD) HR: 1.80; 95%CI: 1.32-2.46). HbA1c was associated with higher risk of pancreatic cancer (per SD HR: 1.21 95%CI 1.05-1.40) and with some suggestion of higher risks for all-cancers-of-interest (per SD HR: 1.05; 95%CI: 0.99-1.11) and colorectal (per SD HR: 1.09; 95%CI: 0.98-1.20), ovarian (per SD HR: 1.18; 95%CI 0.96-1.45) and bladder (per SD HR: 1.08; 95%CI 0.96-1.21) cancers. Compared to no self-reported T2DM and HbA1c <6.5% (reference group), self-reported T2DM and HbA1c <6.5% (i.e., T2DM in good glycemic control) was not associated with risk of colorectal cancer, whereas it was associated with higher risks of all-cancers-of-interest combined (HR: 1.28; 95%CI: 1.01-1.62), especially for breast and endometrial cancers. Additional large, prospective studies are needed to further explore the roles of hyperglycemia, hyperinsulinemia, and related metabolic traits with T2DM-associated cancers to better understand the mechanisms underlying the self-reported T2DM-cancer association and to identify persons at higher cancer risk.
Collapse
Affiliation(s)
- Peter T. Campbell
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
- Population Science Department, American Cancer Society (ACS), Atlanta, Georgia
| | - Christina C. Newton
- Population Science Department, American Cancer Society (ACS), Atlanta, Georgia
| | - Eric J. Jacobs
- Population Science Department, American Cancer Society (ACS), Atlanta, Georgia
| | | | - Ying Wang
- Population Science Department, American Cancer Society (ACS), Atlanta, Georgia
| | - Erika Rees-Punia
- Population Science Department, American Cancer Society (ACS), Atlanta, Georgia
| | - Mark A. Guinter
- Population Science Department, American Cancer Society (ACS), Atlanta, Georgia
| | - Neil Murphy
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC), Lyon, France
| | - Jill Koshiol
- Division of Cancer Epidemiology and Genetics, NIH, NCI, Rockville, Maryland
| | - Ahmed N. Dehal
- Department of Clinical Science, Kaiser Permanente Bernard J Tyson School of Medicine, Panorama City, California
| | - Thomas Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Howard Strickler
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Jessica Petrick
- Slone Epidemiology Center at Boston University, Boston, Massachusetts
| | - Marc Gunter
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC), Lyon, France
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Michael Pollak
- Depsartment of Medicine and Oncology, McGill University, Montreal, Quebec, Canada
| | - Alpa V. Patel
- Population Science Department, American Cancer Society (ACS), Atlanta, Georgia
| | - Susan M. Gapstur
- Population Science Department, American Cancer Society (ACS), Atlanta, Georgia
| |
Collapse
|
12
|
Kumar S, Santos RJ, McGuigan AJ, Singh U, Johnson P, Kunzmann AT, Turkington RC. The Role of Circulating Protein and Metabolite Biomarkers in the Development of Pancreatic Ductal Adenocarcinoma (PDAC): A Systematic Review and Meta-analysis. Cancer Epidemiol Biomarkers Prev 2022; 31:1090-1102. [PMID: 34810209 PMCID: PMC9377754 DOI: 10.1158/1055-9965.epi-21-0616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/19/2021] [Accepted: 11/08/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis, and this is attributed to it being diagnosed at an advanced stage. Understanding the pathways involved in initial development may improve early detection strategies. This systematic review assessed the association between circulating protein and metabolite biomarkers and PDAC development. METHODS A literature search until August 2020 in MEDLINE, EMBASE, and Web of Science was performed. Studies were included if they assessed circulating blood, urine, or salivary biomarkers and their association with PDAC risk. Quality was assessed using the Newcastle-Ottawa scale for cohort studies. Random-effects meta-analyses were used to calculate pooled relative risk. RESULTS A total of 65 studies were included. Higher levels of glucose were found to be positively associated with risk of developing PDAC [n = 4 studies; pooled relative risk (RR): 1.61; 95% CI: 1.16-2.22]. Additionally, an inverse association was seen with pyridoxal 5'-phosphate (PLP) levels (n = 4 studies; RR: 0.62; 95% CI: 0.44-0.87). Meta-analyses showed no association between levels of C-peptide, members of the insulin growth factor signaling pathway, C-reactive protein, adiponectin, 25-hydroxyvitamin D, and folate/homocysteine and PDAC risk. Four individual studies also reported a suggestive positive association of branched-chain amino acids with PDAC risk, but due to differences in measures reported, a meta-analysis could not be performed. CONCLUSIONS Our pooled analysis demonstrates that higher serum glucose levels and lower levels of PLP are associated with risk of PDAC. IMPACT Glucose and PLP levels are associated with PDAC risk. More prospective studies are required to identify biomarkers for early detection.
Collapse
Affiliation(s)
- Swati Kumar
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Ralph J. Santos
- Centre for Public Health, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Andrew J. McGuigan
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Urvashi Singh
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Peter Johnson
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Andrew T. Kunzmann
- Centre for Public Health, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Richard C. Turkington
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| |
Collapse
|
13
|
Zhang X, Wang M, Wang X, Zhu Z, Zhang W, Zhou Z, Tang W, Li Q. Comparison of New Glucose-Lowering Drugs on Risk of Pancreatitis in Type 2 Diabetes: A Network Meta-Analysis. Endocr Pract 2021; 28:333-341. [PMID: 34922031 DOI: 10.1016/j.eprac.2021.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND & AIMS Whether new glucose-lowering drugs increases the risk of pancreatitis in individuals with type 2 diabetes remains controversial. This present network meta-analysis aims to investigate the risk of pancreatitis associated with the use of glucagon-like peptide (GLP)-1 agonists and dipeptidyl peptidase (DPP)-4 inhibitors in the treatment of type 2 diabetes mellitus. METHODS PubMed, Web of Science, EMBASE and the Cochrane Library were searched. The literature was published from the date of their inception to July 21, 2021, including placebo-controlled or head-to-head trials of 2 new glucose-lowering drugs. Relative ratio (RR) and 95% confidence interval (CI) were used to assess the risk of GLP-1 agonists and DPP-4 inhibitors for pancreatitis or pancreatic cancer among patients with type 2 diabetes. RESULTS Seventeen studies were identified, covered 102257 participants. The pooled results showed a neutral relationship between GLP-1 agonists and pancreatitis (Overall RR, 0.96; 95% CI, 0.31-3.00) or pancreatic cancer (Overall RR, 1.10; 95% CI, 0.31-4.10) compared to placebo. Meanwhile, DPP-4 inhibitors were not be associated with the increased risk of pancreatitis (Overall RR, 1.60; 95% CI, 0.25-11.00) or pancreatic cancer (Overall RR, 0.79; 95% CI, 0.26-2.40). Among them, lixisenatide and saxagliptin may be the safest drug compared to other drugs according to the ranking of probability. Sensitivity and subgroup analysis confirmed the stability of the core results. CONCLUSION The most obvious finding to emerge from this study is that GLP-1 agonists and DPP-4 inhibitors are safe with respect to pancreatitis and pancreatic cancer risk compared to placebo. This conclusion may provide useful evidence for correlated clinical researches.
Collapse
Affiliation(s)
- Xuexue Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Department of Endocrinology, China Academy of Chinese Medical Sciences, Beijing, China
| | - Miaoran Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Department of Endocrinology, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xujie Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Department of Endocrinology, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhengchuan Zhu
- Peking University Traditional Chinese Medicine Clinical Medical School [Xi yuan], Beijing, China.
| | - Wantong Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhongyang Zhou
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei Tang
- Department of Endocrinology, Beijing University of Chinese Medicine, Beijing, China
| | - Qiuyan Li
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
14
|
Silke J, O’Reilly LA. NF-κB and Pancreatic Cancer; Chapter and Verse. Cancers (Basel) 2021; 13:4510. [PMID: 34572737 PMCID: PMC8469693 DOI: 10.3390/cancers13184510] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is one of the world's most lethal cancers. An increase in occurrence, coupled with, presently limited treatment options, necessitates the pursuit of new therapeutic approaches. Many human cancers, including PDAC are initiated by unresolved inflammation. The transcription factor NF-κB coordinates many signals that drive cellular activation and proliferation during immunity but also those involved in inflammation and autophagy which may instigate tumorigenesis. It is not surprising therefore, that activation of canonical and non-canonical NF-κB pathways is increasingly recognized as an important driver of pancreatic injury, progression to tumorigenesis and drug resistance. Paradoxically, NF-κB dysregulation has also been shown to inhibit pancreatic inflammation and pancreatic cancer, depending on the context. A pro-oncogenic or pro-suppressive role for individual components of the NF-κB pathway appears to be cell type, microenvironment and even stage dependent. This review provides an outline of NF-κB signaling, focusing on the role of the various NF-κB family members in the evolving inflammatory PDAC microenvironment. Finally, we discuss pharmacological control of NF-κB to curb inflammation, focussing on novel anti-cancer agents which reinstate the process of cancer cell death, the Smac mimetics and their pre-clinical and early clinical trials.
Collapse
Affiliation(s)
- John Silke
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC 3052, Australia;
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Lorraine Ann O’Reilly
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC 3052, Australia;
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
15
|
Duan X, Wang W, Pan Q, Guo L. Type 2 Diabetes Mellitus Intersects With Pancreatic Cancer Diagnosis and Development. Front Oncol 2021; 11:730038. [PMID: 34485159 PMCID: PMC8415500 DOI: 10.3389/fonc.2021.730038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
The relationship between type 2 diabetes mellitus (T2DM) and pancreatic cancer (PC) is complex. Diabetes is a known risk factor for PC, and new-onset diabetes (NOD) could be an early manifestation of PC that may be facilitate the early diagnosis of PC. Metformin offers a clear benefit of inhibiting PC, whereas insulin therapy may increase the risk of PC development. No evidence has shown that novel hypoglycemic drugs help or prevent PC. In this review, the effects of T2DM on PC development are summarized, and novel strategies for the prevention and treatment of T2DM and PC are discussed.
Collapse
Affiliation(s)
- Xiaoye Duan
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Weihao Wang
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Qi Pan
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Lixin Guo
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
16
|
Schorr F, Essig MW. [Early detection of pancreatic cancer - The role of endoscopic and transabdominal ultrasound]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2021; 59:1083-1090. [PMID: 34243212 DOI: 10.1055/a-1515-3123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pancreatic neoplasms are less common tumors and have a poor prognosis when advanced. Early diagnosis would be beneficial for survival, but screening of the whole population cannot be justified with a satisfying benefit-effort correlation. Subgroups of patients with a higher than average risk are those with germ-like mutations, familial cancers risks, and mucinous cystadenomas that would benefit from surveillance programs. Other risk groups, like patients with pancreatitis, new onset diabetes, and heavy smokers, should be considered as well. Transabdominal ultrasonography is of great advantage as a first-line imaging method because of its easy access. The accuracy for adenocarcinoma diagnosis is nearly 90% while using CEUS. Endosonography is of extraordinary importance in the diagnostic approach of pancreatic tumors because of high sensitivity and specificity while using advanced imaging techniques like CEUS, elastography, and fine needle biopsy. Screening by means of EUS is also possible in high-risk situations, and a favourable cost-benefit ratio must be shown by future data.
Collapse
Affiliation(s)
- Friedrich Schorr
- Gastroenterologie - Division Stadtspital/Landspitäler, Insel Gruppe AG, Bern, Switzerland
| | - Manfred Walter Essig
- Gastroenterologie - Division Stadtspital/Landspitäler, Insel Gruppe AG, Bern, Switzerland
| |
Collapse
|
17
|
Pancreatic cancer epidemiology: understanding the role of lifestyle and inherited risk factors. Nat Rev Gastroenterol Hepatol 2021; 18:493-502. [PMID: 34002083 PMCID: PMC9265847 DOI: 10.1038/s41575-021-00457-x] [Citation(s) in RCA: 458] [Impact Index Per Article: 152.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/20/2021] [Indexed: 12/24/2022]
Abstract
Pancreatic cancer is a leading cause of cancer death worldwide and its global burden has more than doubled over the past 25 years. The highest incidence regions for pancreatic cancer include North America, Europe and Australia, and although much of this increase is due to ageing worldwide populations, there are key modifiable risk factors for pancreatic cancer such as cigarette smoking, obesity, diabetes and alcohol intake. The prevalence of these risk factors is increasing in many global regions, resulting in increasing age-adjusted incidence rates for pancreatic cancer, but the relative contribution from these risk factors varies globally due to variation in the underlying prevalence and prevention strategies. Inherited genetic factors, although not directly modifiable, are an important component of pancreatic cancer risk, and include pathogenic variants in hereditary cancer genes, genes associated with hereditary pancreatitis, as well as common variants identified in genome-wide association studies. Identification of the genetic changes that underlie pancreatic cancer not only provides insight into the aetiology of this cancer but also provides an opportunity to guide early detection strategies. The goal of this Review is to provide an up-to-date overview of the established modifiable and inherited risk factors for pancreatic cancer.
Collapse
|
18
|
Molina-Montes E, Coscia C, Gómez-Rubio P, Fernández A, Boenink R, Rava M, Márquez M, Molero X, Löhr M, Sharp L, Michalski CW, Farré A, Perea J, O'Rorke M, Greenhalf W, Iglesias M, Tardón A, Gress TM, Barberá VM, Crnogorac-Jurcevic T, Muñoz-Bellvís L, Dominguez-Muñoz JE, Renz H, Balcells J, Costello E, Ilzarbe L, Kleeff J, Kong B, Mora J, O'Driscoll D, Poves I, Scarpa A, Yu J, Hidalgo M, Lawlor RT, Ye W, Carrato A, Real FX, Malats N. Deciphering the complex interplay between pancreatic cancer, diabetes mellitus subtypes and obesity/BMI through causal inference and mediation analyses. Gut 2021; 70:319-329. [PMID: 32409590 DOI: 10.1136/gutjnl-2019-319990] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 04/07/2020] [Accepted: 04/12/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To characterise the association between type 2 diabetes mellitus (T2DM) subtypes (new-onset T2DM (NODM) or long-standing T2DM (LSDM)) and pancreatic cancer (PC) risk, to explore the direction of causation through Mendelian randomisation (MR) analysis and to assess the mediation role of body mass index (BMI). DESIGN Information about T2DM and related factors was collected from 2018 PC cases and 1540 controls from the PanGenEU (European Study into Digestive Illnesses and Genetics) study. A subset of PC cases and controls had glycated haemoglobin, C-peptide and genotype data. Multivariate logistic regression models were applied to derive ORs and 95% CIs. T2DM and PC-related single nucleotide polymorphism (SNP) were used as instrumental variables (IVs) in bidirectional MR analysis to test for two-way causal associations between PC, NODM and LSDM. Indirect and direct effects of the BMI-T2DM-PC association were further explored using mediation analysis. RESULTS T2DM was associated with an increased PC risk when compared with non-T2DM (OR=2.50; 95% CI: 2.05 to 3.05), the risk being greater for NODM (OR=6.39; 95% CI: 4.18 to 9.78) and insulin users (OR=3.69; 95% CI: 2.80 to 4.86). The causal association between T2DM (57-SNP IV) and PC was not statistically significant (ORLSDM=1.08, 95% CI: 0.86 to 1.29, ORNODM=1.06, 95% CI: 0.95 to 1.17). In contrast, there was a causal association between PC (40-SNP IV) and NODM (OR=2.85; 95% CI: 2.04 to 3.98), although genetic pleiotropy was present (MR-Egger: p value=0.03). Potential mediating effects of BMI (125-SNPs as IV), particularly in terms of weight loss, were evidenced on the NODM-PC association (indirect effect for BMI in previous years=0.55). CONCLUSION Findings of this study do not support a causal effect of LSDM on PC, but suggest that PC causes NODM. The interplay between obesity, PC and T2DM is complex.
Collapse
Affiliation(s)
- Esther Molina-Montes
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain.,CIBERONC, Madrid, Spain
| | - Claudia Coscia
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain.,CIBERONC, Madrid, Spain
| | - Paulina Gómez-Rubio
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain.,CIBERONC, Madrid, Spain
| | - Alba Fernández
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain
| | - Rianne Boenink
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain
| | - Marta Rava
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain
| | - Mirari Márquez
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain.,CIBERONC, Madrid, Spain
| | - Xavier Molero
- Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Barcelona, Spain.,Universitat Autònoma de Barcelona, CIBEREHD, Barcelona, Spain
| | - Matthias Löhr
- Gastrocentrum, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Linda Sharp
- National Cancer Registry Ireland, Cork, Ireland.,HRB Clinical Research Facility, University College Cork, Cork, Ireland
| | - Christoph W Michalski
- Department of Surgery, Technical University of Munich, Munich, Germany.,Department of Visceral, Vascular and Endocrine Surgery, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| | - Antoni Farré
- Department of Gastroenterology and Clinical Biochemistry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - José Perea
- Department of Surgery, Hospital 12 de Octubre, Madrid, Spain.,Department of Surgery, Health Research Institute, Fundación Jiménez Díaz, Madrid, Spain
| | - Michael O'Rorke
- Centre for Public Health, Queen's University Belfast, Belfast, UK.,College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | - William Greenhalf
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
| | - Mar Iglesias
- CIBERONC, Madrid, Spain.,Hospital del Mar-Parc de Salut Mar, Barcelona, Spain
| | - Adonina Tardón
- Department of Medicine, Instituto Universitario de Oncología del Principado de Asturias, Oviedo, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Thomas M Gress
- Department of Gastroenterology, University Hospital of Giessen and Marburg, Marburg, Germany
| | - Victor M Barberá
- Molecular Genetics Laboratory, University General Hospital of Elche, Elche, Spain
| | | | - Luis Muñoz-Bellvís
- Department of Surgery, Hospital Universitario de Salamanca, Salamanca, Spain
| | - J Enrique Dominguez-Muñoz
- Department of Gastroenterology, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Harald Renz
- Institute of Laboratory Medicine and Pathobiochemistry, Phillips University of Marburg, Marburg, Germany
| | - Joaquim Balcells
- Universitat Autònoma de Barcelona, CIBEREHD, Barcelona, Spain.,Exocrine Pancreas Research Unit, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Barcelona, Spain
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
| | - Lucas Ilzarbe
- Department of Gastroenterology, Hospital del Mar - Parc de Salut Mar, Barcelona, Spain
| | - Jörg Kleeff
- Department of Surgery, Technical University of Munich, Munich, Germany.,Department of Visceral, Vascular and Endocrine Surgery, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| | - Bo Kong
- Department of Surgery, Technical University of Munich, Munich, Germany
| | - Josefina Mora
- Department of Gastroenterology and Clinical Biochemistry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Damian O'Driscoll
- National Cancer Registry Ireland, Cork, Ireland.,HRB Clinical Research Facility, University College Cork, Cork, Ireland
| | - Ignasi Poves
- Hospital del Mar - Parc de Salut Mar, Barcelona, Spain
| | - Aldo Scarpa
- ARC-Net Research Centre and Department of Diagnostics and Public Health-Section of Pathology, University of Verona, Verona, Italy
| | - Jingru Yu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Manuel Hidalgo
- Medical Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.,Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| | - Rita T Lawlor
- ARC-Net Research Centre and Department of Diagnostics and Public Health-Section of Pathology, University of Verona, Verona, Italy
| | - Weimin Ye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Alfredo Carrato
- CIBERONC, Madrid, Spain.,Department of Medical Oncology, Ramón y Cajal University Hospital, Madrid, Spain
| | - Francisco X Real
- CIBERONC, Madrid, Spain.,Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center, Madrid, Spain .,CIBERONC, Madrid, Spain
| | | |
Collapse
|
19
|
Kim NH, Chang Y, Lee SR, Ryu S, Kim HJ. Glycemic Status, Insulin Resistance, and Risk of Pancreatic Cancer Mortality in Individuals With and Without Diabetes. Am J Gastroenterol 2020; 115:1840-1848. [PMID: 33156103 DOI: 10.14309/ajg.0000000000000956] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The impact of glycemic status and insulin resistance on the risk of pancreatic cancer in the nondiabetic population remains uncertain. We aimed to examine the association of glycemic status and insulin resistance with pancreatic cancer mortality in individuals with and without diabetes. METHODS This is a cohort study of 572,021 Korean adults without cancer at baseline, who participated in repeat screening examinations which included fasting blood glucose, hemoglobin A1c, and insulin, and were followed for a median of 8.4 years (interquartile range, 5.3 -13.2 years). Vital status and pancreatic cancer mortality were ascertained through linkage to national death records. RESULTS During 5,211,294 person-years of follow-up, 260 deaths from pancreatic cancer were identified, with a mortality rate of 5.0 per 10 person-years. In the overall population, the risk of pancreatic cancer mortality increased with increasing levels of glucose and hemoglobin A1c in a dose-response manner, and this association was observed even in individuals without diabetes. In nondiabetic individuals without previously diagnosed or screen-detected diabetes, insulin resistance and hyperinsulinemia were positively associated with increased pancreatic cancer mortality. Specifically, the multivariable-adjusted hazard ratio (95% confidence intervals) for pancreatic cancer mortality comparing the homeostatic model assessment of insulin resistance ≥75th percentile to the <75th percentile was 1.49 (1.08-2.05), and the corresponding hazard ratio comparing the insulin ≥75th percentile to the <75th percentile was 1.43 (1.05-1.95). These associations remained significant when introducing changes in insulin resistance, hyperinsulinemia, and other confounders during follow-up as time-varying covariates. DISCUSSION Glycemic status, insulin resistance, and hyperinsulinemia, even in individuals without diabetes, were independently associated with an increased risk of pancreatic cancer mortality.
Collapse
Affiliation(s)
- Nam Hee Kim
- Division of Gastroenterology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yoosoo Chang
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Sung Ryol Lee
- Department of Surgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seungho Ryu
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Hong Joo Kim
- Division of Gastroenterology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
20
|
Diabetes und Krebs. DIABETOLOGE 2020. [DOI: 10.1007/s11428-020-00677-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
21
|
Abstract
OBJECTIVES Type 2 diabetes mellitus (T2DM) has been associated with several types of cancers, but the role of T2DM in pancreatic neuroendocrine tumors (pNETs) has not been systematically studied. METHODS In this study, 299 patients with pNETs were recruited, and the clinicopathologic characteristics and prognosis of the diabetic and nondiabetic patients were compared. The association between metformin use and survival was assessed to examine whether metformin impacts the prognosis of pNETs patients. RESULTS The prevalence of T2DM in the cohort was 20.7% (n = 62). The proportions of grade 3 tumors, distant metastases, and nerve invasion in pNET patients with T2DM were higher than those in patients without T2DM, and as a result, the survival was worse in patients with T2DM. After adjusting for the tumor stage, diabetic status was not associated with poor survival in the univariate analysis. The results of logistic regression showed that pNET patients with T2DM were at high risk for tumor metastasis (odds ratio [OR], 2.81; P = 0.001), nerve invasion (OR, 2.43; P = 0.029), and grade 3 tumors (OR, 4.97; P = 0.010). CONCLUSIONS Type 2 diabetes mellitus is associated with pNET metastasis and not an independent risk factor for poor prognosis in pNETs.
Collapse
|
22
|
Lin Y, Nakatochi M, Hosono Y, Ito H, Kamatani Y, Inoko A, Sakamoto H, Kinoshita F, Kobayashi Y, Ishii H, Ozaka M, Sasaki T, Matsuyama M, Sasahira N, Morimoto M, Kobayashi S, Fukushima T, Ueno M, Ohkawa S, Egawa N, Kuruma S, Mori M, Nakao H, Adachi Y, Okuda M, Osaki T, Kamiya S, Wang C, Hara K, Shimizu Y, Miyamoto T, Hayashi Y, Ebi H, Kohmoto T, Imoto I, Kasugai Y, Murakami Y, Akiyama M, Ishigaki K, Matsuda K, Hirata M, Shimada K, Okusaka T, Kawaguchi T, Takahashi M, Watanabe Y, Kuriki K, Kadota A, Okada R, Mikami H, Takezaki T, Suzuki S, Yamaji T, Iwasaki M, Sawada N, Goto A, Kinoshita K, Fuse N, Katsuoka F, Shimizu A, Nishizuka SS, Tanno K, Suzuki K, Okada Y, Horikoshi M, Yamauchi T, Kadowaki T, Yu H, Zhong J, Amundadottir LT, Doki Y, Ishii H, Eguchi H, Bogumil D, Haiman CA, Le Marchand L, Mori M, Risch H, Setiawan VW, Tsugane S, Wakai K, Yoshida T, Matsuda F, Kubo M, Kikuchi S, Matsuo K. Genome-wide association meta-analysis identifies GP2 gene risk variants for pancreatic cancer. Nat Commun 2020; 11:3175. [PMID: 32581250 PMCID: PMC7314803 DOI: 10.1038/s41467-020-16711-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 05/15/2020] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer is the fourth leading cause of cancer-related deaths in Japan. To identify risk loci, we perform a meta-analysis of three genome-wide association studies comprising 2,039 pancreatic cancer patients and 32,592 controls in the Japanese population. Here, we identify 3 (13q12.2, 13q22.1, and 16p12.3) genome-wide significant loci (P < 5.0 × 10−8), of which 16p12.3 has not been reported in the Western population. The lead single nucleotide polymorphism (SNP) at 16p12.3 is rs78193826 (odds ratio = 1.46, 95% confidence interval = 1.29-1.66, P = 4.28 × 10−9), an Asian-specific, nonsynonymous glycoprotein 2 (GP2) gene variant. Associations between selected GP2 gene variants and pancreatic cancer are replicated in 10,822 additional cases and controls of East Asian origin. Functional analyses using cell lines provide supporting evidence of the effect of rs78193826 on KRAS activity. These findings suggest that GP2 gene variants are probably associated with pancreatic cancer susceptibility in populations of East Asian ancestry. Previous genome-wide association studies have identified risk loci for pancreatic cancer but were centered on individuals of European ancestry. Here the authors identify GP2 gene variants associated with pancreatic cancer susceptibility in populations of East Asian ancestry.
Collapse
Affiliation(s)
- Yingsong Lin
- Department of Public Health, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan.
| | - Masahiro Nakatochi
- Division of Public Health Informatics, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, 461-8673, Japan. .,Department of Nursing, Nagoya University Graduate School of Medicine, Nagoya, 461-8673, Japan.
| | - Yasuyuki Hosono
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan
| | - Hidemi Ito
- Division of Cancer Information and Control, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan.,Department of Descriptive Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.,Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Akihito Inoko
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan.,Department of Pathology, Aichi Medical University School of Medicine, Nagakute, 480-1195, Japan
| | - Hiromi Sakamoto
- Genetics Division, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Fumie Kinoshita
- Data Science Division, Data Coordinating Center, Department of Advanced Medicine, Nagoya University Hospital, Nagoya, 461-8673, Japan
| | - Yumiko Kobayashi
- Data Science Division, Data Coordinating Center, Department of Advanced Medicine, Nagoya University Hospital, Nagoya, 461-8673, Japan
| | | | - Masato Ozaka
- Department of Hepato-biliary-pancreatic Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Takashi Sasaki
- Department of Hepato-biliary-pancreatic Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Masato Matsuyama
- Department of Hepato-biliary-pancreatic Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Naoki Sasahira
- Department of Hepato-biliary-pancreatic Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Manabu Morimoto
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Yokohama, 241-8515, Japan
| | - Satoshi Kobayashi
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Yokohama, 241-8515, Japan
| | - Taito Fukushima
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Yokohama, 241-8515, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Yokohama, 241-8515, Japan
| | - Shinichi Ohkawa
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Yokohama, 241-8515, Japan
| | - Naoto Egawa
- Department of Gastroenterology, Tokyo Metropolitan Hiroo Hospital, Tokyo, 150-0013, Japan
| | - Sawako Kuruma
- Department of Gastroenterology, Tokyo Metropolitan Komagome Hospital, Tokyo, 113-8677, Japan
| | - Mitsuru Mori
- Hokkaido Chitose College of Rehabilitation, Hokkaido, 066-0055, Japan
| | - Haruhisa Nakao
- Division of Hepatology and Pancreatology, Aichi Medical University School of Medicine, Nagakute, 480-1195, Japan
| | | | - Masumi Okuda
- Department of Pediatrics, Hyogo College of Medicine, Nishinomiya, Hyogo, 663-8501, Japan
| | - Takako Osaki
- Department of Infectious Diseases, Kyorin University School of Medicine, Tokyo, 181-8611, Japan
| | - Shigeru Kamiya
- Department of Infectious Diseases, Kyorin University School of Medicine, Tokyo, 181-8611, Japan
| | - Chaochen Wang
- Department of Public Health, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Kazuo Hara
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, 464-8681, Japan
| | - Yasuhiro Shimizu
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Nagoya, 464-8681, Japan
| | - Tatsuo Miyamoto
- Department of Genetics and Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Yuko Hayashi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan
| | - Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan
| | - Tomohiro Kohmoto
- Department of Human Genetics, Tokushima University Graduate School of Medicine, Tokushima, 770-8503, Japan.,Division of Molecular Genetics, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan
| | - Issei Imoto
- Division of Molecular Genetics, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan
| | - Yumiko Kasugai
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan.,Department of Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Yoshinori Murakami
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Masato Akiyama
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.,Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kazuyoshi Ishigaki
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Koichi Matsuda
- Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Makoto Hirata
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Kazuaki Shimada
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Meiko Takahashi
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Yoshiyuki Watanabe
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Kiyonori Kuriki
- Laboratory of Public Health, School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Aya Kadota
- Department of Public Health, Shiga University of Medical Science, Otsu, 520-2192, Japan
| | - Rieko Okada
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Haruo Mikami
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, 260-8717, Japan
| | - Toshiro Takezaki
- Department of International Island and Community Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8544, Japan
| | - Sadao Suzuki
- Department of Public Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Taiki Yamaji
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, 104-0045, Japan
| | - Motoki Iwasaki
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, 104-0045, Japan
| | - Norie Sawada
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, 104-0045, Japan
| | - Atsushi Goto
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, 104-0045, Japan
| | - Kengo Kinoshita
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, 980-8573, Japan
| | - Nobuo Fuse
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, 980-8573, Japan
| | - Fumiki Katsuoka
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, 980-8573, Japan
| | - Atsushi Shimizu
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, 028-3694, Japan
| | - Satoshi S Nishizuka
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, 028-3694, Japan
| | - Kozo Tanno
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, 028-3694, Japan.,Department of Hygiene and Preventive Medicine, School of Medicine, Iwate Medicalm University, Iwate, 028-3694, Japan
| | - Ken Suzuki
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.,Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.,Laboratory for Endocrinology, Metabolism and Kidney Diseases, RIKEN Centre for Integrative Medical Sciences, Yokohama, 230-0045, Japan.,Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Yukinori Okada
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.,Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan.,Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka, 565-0871, Japan
| | - Momoko Horikoshi
- Laboratory for Endocrinology, Metabolism and Kidney Diseases, RIKEN Centre for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Herbert Yu
- University of Hawaii Cancer Center, Honolulu, HI, 96813, USA
| | - Jun Zhong
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Hideshi Ishii
- Department of Medical Data Science, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - David Bogumil
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeless, CA, 90033, USA
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeless, CA, 90033, USA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA
| | | | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Harvey Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, 06520, USA
| | - Veronica W Setiawan
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeless, CA, 90033, USA.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA
| | - Shoichiro Tsugane
- Center for Public Health Sciences, National Cancer Center, Tokyo, 104-0045, Japan
| | - Kenji Wakai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Teruhiko Yoshida
- Genetics Division, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Shogo Kikuchi
- Department of Public Health, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan. .,Department of Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| |
Collapse
|
23
|
Abstract
PURPOSE Pancreatic cancer is the seventh cause of death in men in the world and also is one of the most common malignancies worldwide with poor prognosis. Due to the lack of epidemiological aspects of pancreatic cancer in Iran, this study aimed to investigate the epidemiological aspects of pancreatic cancer in Iran. METHODS In February 2019, A search was conducted with the keywords "Pancreatic Neoplasm," "Pancreas Neoplasms," "Cancer of Pancreas," "Pancreas Cancer," "Pancreatic Cancer," "Cancer of the Pancreas," and "Iran" in their title or abstract and MeSH. The databases of Medline, Web of Science, Scopus, SID, IranMedex, and Google Scholar were searched. The title and abstract of the papers were reviewed, and articles that addressed the epidemiological aspects of pancreatic cancer were included; in total, 20 full papers were reviewed. RESULTS According to studies, the incidence and mortality rate of pancreatic cancer is steadily increasing with age. Actually, men are more likely to develop this carcinoma than women. Smoking, aging, and lifestyle changes are the most important risk factors for pancreatic cancer in Iran. Due to the lack of initial symptoms or a specific marker for early diagnosis of pancreatic cancer, this cancer is detected lately and therefore low survival rate is observed. CONCLUSION Mortality and incidence of pancreatic cancer is increasing in Iran. Pancreatic cancer is more common in men than women. Based on the results of this study, pancreatic cancer depends largely on the lifestyle. Survival of pancreatic cancer is low in untreated patients.
Collapse
Affiliation(s)
| | - Hamid Salehiniya
- Social Determinants of Health Research Center, Birjand University of Medical Sciences, Birjand, Iran.
- Department of Epidemiology and Biostatistics, Tehran University of medical sciences, Tehran, Iran.
| |
Collapse
|
24
|
Peila R, Rohan TE. Diabetes, Glycated Hemoglobin, and Risk of Cancer in the UK Biobank Study. Cancer Epidemiol Biomarkers Prev 2020; 29:1107-1119. [PMID: 32179703 DOI: 10.1158/1055-9965.epi-19-1623] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/24/2020] [Accepted: 03/06/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Evidence suggest that diabetes and glycated hemoglobin (HbA1c) levels are associated with cancer risk. However, previous studies have been limited variably by failure to adjust for cancer-specific risk factors (e.g., body mass index), inattention to diabetes duration and use of antidiabetic medications, and failure to stratify by obesity. METHODS We examined the association between diabetes, HbA1c, and cancer risk in the UK Biobank, using data from 476,517 participants (54% women), followed for an average period of 7.1 years. Diabetes was defined on the basis of baseline self-reported diagnosis of diabetes and/or use of diabetes medication, while HbA1c measured at baseline was categorized as low (<31 mmol/mol), normal (31-<39 mmol/mol), increased risk (39-<48 mmol/mol), and high risk for diabetes (≥48 mmol/mol). Multivariable Cox proportional hazards models were used to estimate the association of diabetes and cancer at different anatomical sites, with adjustment for cancer-specific risk factors. RESULTS Diabetes was associated with increased risk of cancers of the stomach, liver, bladder, endometrium, and lung among smokers, and with decreased risk of prostate cancer. Compared with the normal HbA1c category, the increased risk category was positively associated with risk of cancers of the colon, liver, bladder, and lung among smokers, and the high-risk category was associated with increased risk of cancers of the esophagus, liver, pancreas, and bladder, and with decreased risk of prostate cancer. CONCLUSIONS These results suggest that both diabetes and/or elevated HbA1c are associated with risk of cancer at several anatomic sites. IMPACT The associations of diabetes and HbA1c levels with cancer suggest their importance in cancer prevention.
Collapse
Affiliation(s)
- Rita Peila
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York.
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
25
|
Zhou B, Wu D, Liu H, Du LT, Wang YS, Xu JW, Qiu FB, Hu SY, Zhan HX. Obesity and pancreatic cancer: An update of epidemiological evidence and molecular mechanisms. Pancreatology 2019; 19:941-950. [PMID: 31447281 DOI: 10.1016/j.pan.2019.08.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 07/04/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022]
Abstract
Despite advances in therapy and achievements in translational research, pancreatic cancer (PC) remains an invariably fatal malignancy. Risk factors that affect the incidence of PC include diabetes, smoking, obesity, chronic pancreatitis, and diet. The growing worldwide obesity epidemic is associated with an increased risk of the most common cancers, including PC. Chronic inflammation, hormonal effects, circulating adipokines, and adipocyte-mediated inflammatory and immunosuppressive microenvironment are involved in the association of obesity with PC. Herein, we systematically review the epidemiology of PC and the biological mechanisms that may account for this association. Included in this review is a discussion of adipokine-mediated inflammation, lipid metabolism, and the interactions of adipocytes with cancer cells. We consider the influence of bariatric surgery on the risk of PC risk as well as potential molecular targets of therapy. Our review leads us to conclude that targeting adipose tissue to achieve weight loss may represent a new therapeutic strategy for preventing and treating PC.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China; Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China
| | - Dong Wu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Han Liu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Lu-Tao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, 250033, China; Tumor Marker Detection Engineering Laboratory of Shandong Province, Jinan, Shandong Province, 250033, China
| | - Yun-Shan Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong Province, 250033, China; Tumor Marker Detection Engineering Laboratory of Shandong Province, Jinan, Shandong Province, 250033, China
| | - Jian-Wei Xu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Fa-Bo Qiu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China; Department of Retroperitoneal Tumor Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China
| | - San-Yuan Hu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Han-Xiang Zhan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China.
| |
Collapse
|
26
|
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: 4.4] [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
|
27
|
Hidaka A, Budhathoki S, Yamaji T, Sawada N, Tanaka-Mizuno S, Kuchiba A, Charvat H, Goto A, Shimazu T, Inoue M, Noda M, Tsugane S, Iwasaki M. Plasma C-peptide and glycated albumin and subsequent risk of cancer: From a large prospective case-cohort study in Japan. Int J Cancer 2019; 144:718-729. [PMID: 30183080 DOI: 10.1002/ijc.31847] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 01/18/2023]
Abstract
To elucidate the individual impacts of insulin and blood glucose on cancer risk, we investigated the association of plasma C-peptide, a surrogated marker of insulin and glycated albumin (GA), a more stable marker of blood glucose, with all-site and site-specific cancer risk by mutually accounting for their confounding effects. The study was prospectively conducted with nearly 4,000 cancer cases arising in our population-based cohort of 33,736 subjects who answered the baseline questionnaire and supplied blood samples. After exclusion of subjects with apparent DM, analysis was done in 3,036 cancer cases and 3,667 subcohort subjects. Among men and women combined, highest levels of C-peptide were statistically significantly associated with an increased risk of all-site [Hazard ratio (HR): 1.21; 95% confidence interval: 1.02-1.42], colon [1.73; 1.20-2.47], liver [3.23; 1.76-5.91], kidney, renal pelvis and ureter cancers [2.47; 1.07-5.69], compared to the respective lowest levels, after adjustment for GA levels. Among these C-peptide-related cancers, colon and liver cancers also showed an increased risk associated with elevated GA levels independently of C-peptide levels. The corresponding HRs for colon and liver cancers compared to the highest and lowest GA levels were 1.43 [1.02-2.00] and 2.02 [1.15-3.55], respectively. Effect modification by gender was only evident for the association between C-peptide and colon cancer (p for interaction = 0.04). Higher insulin levels, independently of higher blood glucose levels, may be relevant to DM-related carcinogenesis for several cancer sites. Examination of circulating insulin levels is a plausible option in evaluating cancer risk even in individuals who have not developed DM.
Collapse
Affiliation(s)
- Akihisa Hidaka
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Sanjeev Budhathoki
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Taiki Yamaji
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Norie Sawada
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | | | - Aya Kuchiba
- Division of Biostatistical Research, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan.,Biostatistics Division, Center for Research Administration and Support, National Cancer Center, Tokyo, Japan
| | - Hadrien Charvat
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Atsushi Goto
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Taichi Shimazu
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Manami Inoue
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Mitsuhiko Noda
- Department of Endocrinology and Diabetes, Saitama Medical University, Saitama, Japan
| | - Shoichiro Tsugane
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Motoki Iwasaki
- Epidemiology and Prevention Group, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | | |
Collapse
|
28
|
Abstract
Obesity is a risk factor for all major gastrointestinal cancers. With the rapid increase in the prevalence of obesity worldwide, this link could lead to an elevated burden of cancers of the digestive system. Currently, three main mechanisms explaining the link between excess adiposity and gastrointestinal cancer risk are being considered, including altered insulin signaling, obesity-associated chronic low-grade inflammation, and altered sex hormone metabolism, although new potential mechanisms emerge. This review is aimed to present our current knowledge on biological mechanisms involved in adiposity-related gastrointestinal carcinogenesis supported by results collected in epidemiological studies.
Collapse
|
29
|
McGuigan A, Kelly P, Turkington RC, Jones C, Coleman HG, McCain RS. Pancreatic cancer: A review of clinical diagnosis, epidemiology, treatment and outcomes. World J Gastroenterol 2018; 24:4846-4861. [PMID: 30487695 PMCID: PMC6250924 DOI: 10.3748/wjg.v24.i43.4846] [Citation(s) in RCA: 1105] [Impact Index Per Article: 184.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 10/19/2018] [Accepted: 10/27/2018] [Indexed: 02/06/2023] Open
Abstract
This review aims to outline the most up-to-date knowledge of pancreatic adenocarcinoma risk, diagnostics, treatment and outcomes, while identifying gaps that aim to stimulate further research in this understudied malignancy. Pancreatic adenocarcinoma is a lethal condition with a rising incidence, predicted to become the second leading cause of cancer death in some regions. It often presents at an advanced stage, which contributes to poor five-year survival rates of 2%-9%, ranking firmly last amongst all cancer sites in terms of prognostic outcomes for patients. Better understanding of the risk factors and symptoms associated with this disease is essential to inform both health professionals and the general population of potential preventive and/or early detection measures. The identification of high-risk patients who could benefit from screening to detect pre-malignant conditions such as pancreatic intraepithelial neoplasia, intraductal papillary mucinous neoplasms and mucinous cystic neoplasms is urgently required, however an acceptable screening test has yet to be identified. The management of pancreatic adenocarcinoma is evolving, with the introduction of new surgical techniques and medical therapies such as laparoscopic techniques and neo-adjuvant chemoradiotherapy, however this has only led to modest improvements in outcomes. The identification of novel biomarkers is desirable to move towards a precision medicine era, where pancreatic cancer therapy can be tailored to the individual patient, while unnecessary treatments that have negative consequences on quality of life could be prevented for others. Research efforts must also focus on the development of new agents and delivery systems. Overall, considerable progress is required to reduce the burden associated with pancreatic cancer. Recent, renewed efforts to fund large consortia and research into pancreatic adenocarcinoma are welcomed, but further streams will be necessary to facilitate the momentum needed to bring breakthroughs seen for other cancer sites.
Collapse
Affiliation(s)
- Andrew McGuigan
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast BT9 7AE, United Kingdom
| | - Paul Kelly
- Department of Pathology, Royal Victoria Hospital, Belfast BT12 6BA, United Kingdom
| | - Richard C Turkington
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast BT9 7AE, United Kingdom
| | - Claire Jones
- Department of Hepatobiliary Surgery, Mater Hospital, Belfast BT14 6AB, United Kingdom
| | - Helen G Coleman
- Centre for Public Health, Queen’s University Belfast, Belfast BT12 6BJ, United Kingdom
| | - R Stephen McCain
- Department of Hepatobiliary Surgery, Mater Hospital, Belfast BT14 6AB, United Kingdom
- Centre for Public Health, Queen’s University Belfast, Belfast BT12 6BJ, United Kingdom
| |
Collapse
|
30
|
Murphy N, Jenab M, Gunter MJ. Adiposity and gastrointestinal cancers: epidemiology, mechanisms and future directions. Nat Rev Gastroenterol Hepatol 2018; 15:659-670. [PMID: 29970888 DOI: 10.1038/s41575-018-0038-1] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Excess adiposity is a risk factor for several cancers of the gastrointestinal system, specifically oesophageal adenocarcinoma and colorectal, small intestine, pancreatic, liver, gallbladder and stomach cancers. With the increasing prevalence of obesity in nearly all regions of the world, this relationship could represent a growing source of cancers of the digestive system. Experimental and molecular epidemiological studies indicate important roles for alterations in insulin signalling, adipose tissue-derived inflammation and sex hormone pathways in mediating the association between adiposity and gastrointestinal cancer. The intestinal microbiome, gut hormones and non alcoholic fatty liver disease (NAFLD) also have possible roles. However, important gaps remain in our knowledge. For instance, our understanding of how adiposity throughout the life course is related to the risk of gastrointestinal cancer development and of how obesity influences gastrointestinal cancer prognosis and survival is limited. Nonetheless, the increasing use of state-of-the-art analytical methods (such as omics technologies, Mendelian randomization and MRI) in large-scale epidemiological studies offers exciting opportunities to advance our understanding of the complex relationship between adiposity and gastrointestinal cancers. Here, we examine the epidemiology of associations between obesity and gastrointestinal cancer, explore potential mechanisms underlying these relationships and highlight important unanswered research questions.
Collapse
Affiliation(s)
- Neil Murphy
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Mazda Jenab
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Marc J Gunter
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France.
| |
Collapse
|
31
|
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: 2.3] [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
|
32
|
Deng T, Lyon CJ, Bergin S, Caligiuri MA, Hsueh WA. Obesity, Inflammation, and Cancer. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 11:421-49. [PMID: 27193454 DOI: 10.1146/annurev-pathol-012615-044359] [Citation(s) in RCA: 532] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Obesity, a worldwide epidemic, confers increased risk for multiple serious conditions, including cancer, and is increasingly recognized as a growing cause of preventable cancer risk. Chronic inflammation, a well-known mediator of cancer, is a central characteristic of obesity, leading to many of its complications, and obesity-induced inflammation confers additional cancer risk beyond obesity itself. Multiple mechanisms facilitate this strong association between cancer and obesity. Adipose tissue is an important endocrine organ, secreting several hormones, including leptin and adiponectin, and chemokines that can regulate tumor behavior, inflammation, and the tumor microenvironment. Excessive adipose expansion during obesity causes adipose dysfunction and inflammation to increase systemic levels of proinflammatory factors. Cells from adipose tissue, such as cancer-associated adipocytes and adipose-derived stem cells, enter the cancer microenvironment to enhance protumoral effects. Dysregulated metabolism that stems from obesity, including insulin resistance, hyperglycemia, and dyslipidemia, can further impact tumor growth and development. This review describes how adipose tissue becomes inflamed in obesity, summarizes ways these mechanisms impact cancer development, and discusses their role in four adipose-associated cancers that demonstrate elevated incidence or mortality in obesity.
Collapse
Affiliation(s)
- Tuo Deng
- Diabetes Research Center and Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas 77030; .,Department of Medicine, Weill Cornell Medical College at Cornell University, New York, New York 10021
| | - Christopher J Lyon
- Diabetes Research Center and Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas 77030;
| | - Stephen Bergin
- Medical Scientist Training Program and Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio 43210.,The Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio 43210
| | - Michael A Caligiuri
- The Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio 43210
| | - Willa A Hsueh
- The Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The Ohio State University, Columbus, Ohio 43210;
| |
Collapse
|
33
|
Abstract
Pancreatic cancer has few known risk factors, providing little in the way of prevention, and is the most rapidly fatal cancer with 7 % survival rate at 5 years. Obesity has surfaced as an important risk factor for pancreatic cancer as epidemiological studies with strong methodological designs have removed important biases and solidified the obesity associations. Moreover, studies indicate that obesity early in adulthood is strongly associated with future risk of pancreatic cancer and that abdominal obesity is an independent risk factor. There is increasing evidence suggesting long-standing diabetes type 2 and insulin resistance are important etiological factors of this disease, providing a strong mechanistic link to obesity. The challenge remains to determine whether intended weight loss in midlife will reduce risk of pancreatic cancer and to elucidate the complex underlying pathways directly involved with risk.
Collapse
Affiliation(s)
- Dominique S Michaud
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
34
|
Nogueira LM, Newton CC, Pollak M, Silverman DT, Albanes D, Männistö S, Weinstein SJ, Jacobs EJ, Stolzenberg-Solomon RZ. Serum C-peptide, Total and High Molecular Weight Adiponectin, and Pancreatic Cancer: Do Associations Differ by Smoking? Cancer Epidemiol Biomarkers Prev 2017; 26:914-922. [PMID: 28096201 PMCID: PMC5457333 DOI: 10.1158/1055-9965.epi-16-0891] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 12/16/2022] Open
Abstract
Background: Studies examining associations between circulating concentrations of C-peptide and total adiponectin, two biomarkers related to obesity and insulin secretion and sensitivity and pancreatic ductal adenocarcinoma (PDA) risk have shown inconsistent results and included limited numbers of smokers.Methods: We examined associations of these biomarkers and high molecular weight (HMW) adiponectin with PDA, overall, and by smoking status. We conducted a pooled nested case-control analysis in 3 cohorts (Prostate, Lung, Colorectal, and Ovarian Cancer Trial, Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study, and Cancer Prevention Study-II), with 758 cases (435 current smokers) and 1,052 controls (531 smokers) matched by cohort, age, sex, race, blood draw date and follow-up time. We used conditional logistic regression adjusted for age, smoking, diabetes, and body mass index to calculate ORs and 95% confidence intervals (CI).Results: Circulating C-peptide concentration was not associated with PDA in never or former smokers, but was inversely associated with PDA in current smokers (per SD OR = 0.67; 95% CI, 0.54-0.84; Pinteraction = 0.005). HMW adiponectin was inversely associated with PDA in never smokers (OR = 0.43; 95% CI, 0.23-0.81), not associated in former smokers, and positively associated in smokers (OR = 1.23; 95% CI, 1.04-1.45; Pinteraction = 0.009). Total adiponectin was not associated with PDA in nonsmokers or current smokers.Conclusions: Associations of biomarkers of insulin secretion and sensitivity with PDA differ by smoking status. Smoking-induced pancreatic damage may explain the associations in smokers while mechanisms related to insulin resistance associations in nonsmokers.Impact: Future studies of these biomarkers and PDA should examine results by smoking status. Cancer Epidemiol Biomarkers Prev; 26(6); 914-22. ©2017 AACR.
Collapse
Affiliation(s)
- Leticia M Nogueira
- Texas Cancer Registry, Department of State Health Services, Austin, Texas
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, Rockville, Maryland
| | | | - Michael Pollak
- Department of Oncology, Lady Davis Research Institute of the Jewish General Hospital and McGill University, Montreal, Quebec, Canada
| | - Debra T Silverman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, Rockville, Maryland
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, Rockville, Maryland
| | - Satu Männistö
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Stephanie J Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, Rockville, Maryland
| | - Eric J Jacobs
- Epidemiology Research Program, American Cancer Society, Atlanta Georgia.
| | | |
Collapse
|
35
|
Fasting but not casual blood glucose is associated with pancreatic cancer mortality in Japanese: EPOCH-JAPAN. Cancer Causes Control 2017; 28:625-633. [DOI: 10.1007/s10552-017-0884-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 03/11/2017] [Indexed: 12/20/2022]
|
36
|
Pang Y, Kartsonaki C, Guo Y, Bragg F, Yang L, Bian Z, Chen Y, Iona A, Millwood IY, Lv J, Yu C, Chen J, Li L, Holmes MV, Chen Z. Diabetes, plasma glucose and incidence of pancreatic cancer: A prospective study of 0.5 million Chinese adults and a meta-analysis of 22 cohort studies. Int J Cancer 2017; 140:1781-1788. [PMID: 28063165 PMCID: PMC5396360 DOI: 10.1002/ijc.30599] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 11/27/2016] [Accepted: 12/20/2016] [Indexed: 12/12/2022]
Abstract
Diabetes is associated with an increased risk of pancreatic cancer (PC) in Western populations. Uncertainty remains, however, about the relevance of plasma glucose for PC among people without diabetes and about the associations of diabetes and high blood glucose with PC in China where the increase in diabetes prevalence has been very recent. The prospective China Kadoorie Biobank (CKB) study recruited 512,000 adults aged 30‐79 years from 10 diverse areas of China during 2004‐2008, recording 595 PC cases during 8 years of follow‐up. Cox regression yielded adjusted hazard ratios (HRs) for PC associated with diabetes (previously diagnosed or screen‐detected) and, among those without previously diagnosed diabetes, with levels of random plasma glucose (RPG). These were further meta‐analysed with 22 published prospective studies. Overall 5.8% of CKB participants had diabetes at baseline. Diabetes was associated with almost twofold increased risk of PC (adjusted HR = 1.87, 95% CI 1.48‐2.37), with excess risk higher in those with longer duration since diagnosis (p for trend = 0.01). Among those without previously diagnosed diabetes, each 1 mmol/L higher usual RPG was associated with a HR of 1.12 (1.04‐1.21). In meta‐analysis of CKB and 22 other studies, previously diagnosed diabetes was associated with a 52% excess risk (1.52, 1.43‐1.63). Among those without diabetes, each 1 mmol/L higher blood glucose was associated with a 15% (1.15, 1.09‐1.21) excess risk. In Chinese and non‐Chinese populations, diabetes and higher blood glucose levels among those without diabetes are associated with an increased risk of PC. What's new? Diabetes is associated with increased risk of pancreatic cancer (PC) in Western populations, but uncertainty remains about the relevance of plasma glucose among people without diabetes. In this first prospective study investigating the association of diabetes with PC risk in China, where the increase in diabetes prevalence has been very recent, diabetes was associated with an almost twofold increased risk of PC, with random plasma glucose being positively associated with PC risk among participants without prior diagnosis of diabetes. In meta‐analysis, the results were consistent with previous evidence in Western populations, highlighting diabetes as a potential aetiological factor of PC.
Collapse
Affiliation(s)
- Yuanjie Pang
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Christiana Kartsonaki
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.,Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Oxford, United Kingdom
| | - Yu Guo
- Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Fiona Bragg
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Ling Yang
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.,Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Oxford, United Kingdom
| | - Zheng Bian
- Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yiping Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.,Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Oxford, United Kingdom
| | - Andri Iona
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.,Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Oxford, United Kingdom
| | - Iona Y Millwood
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.,Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Oxford, United Kingdom
| | - Jun Lv
- School of Public Health, Peking University, Beijing, 100191, China
| | - Canqing Yu
- School of Public Health, Peking University, Beijing, 100191, China
| | - Junshi Chen
- National Center for Food Safety Risk Assessment, Beijing, 100021, China
| | - Liming Li
- Chinese Academy of Medical Sciences, Beijing, 100730, China.,School of Public Health, Peking University, Beijing, 100191, China
| | - Michael V Holmes
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.,Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Oxford, United Kingdom
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
37
|
Dugnani E, Balzano G, Pasquale V, Scavini M, Aleotti F, Liberati D, Di Terlizzi G, Gandolfi A, Petrella G, Reni M, Doglioni C, Bosi E, Falconi M, Piemonti L. Insulin resistance is associated with the aggressiveness of pancreatic ductal carcinoma. Acta Diabetol 2016; 53:945-956. [PMID: 27552832 DOI: 10.1007/s00592-016-0893-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/27/2016] [Indexed: 12/15/2022]
Abstract
AIMS To study whether insulin resistance accelerates the development and/or the progression of pancreatic adenocarcinoma (PDAC), we hypothesized that patients with insulin resistance, compared with those without insulin resistance, show: (1) a younger age and more advanced PDAC stage at diagnosis and (2) a shorter disease-free and overall survival after PDAC diagnosis. METHODS Prospective observational study of patients admitted to a referral center for pancreatic disease. Insulin resistance was defined as a HOMA-IR value greater than the 66th percentile value of the patients included in this study. Survival was estimated according to Kaplan-Meier and by Cox regression. RESULTS Of 296 patients with PDAC, 99 (33 %) met criteria for being classified as insulin resistant at diagnosis. Median follow-up time after diagnosis was 5.27 ± 0.23 years. Patients with insulin resistance received a diagnosis of PDAC at a similar age compared to patients without insulin resistance (67.1 ± 9 vs. 66.8 ± 10 years, p = 0.68), but were more likely to have a cancer stage ≥3 (23.2 vs. 14.2 %, p = 0.053) and a residual disease after surgery (R1 56.4 vs. 38 %; p = 0.007). The median overall survival was 1.3 ± 0.14 and 1.79 ± 0.11 years for the patients with and without insulin resistance, respectively (p = 0.016). Results did not change when patients with diabetes at PDAC diagnosis were excluded from the analysis. Multivariate analysis showed that insulin resistance was independently associated with overall survival. CONCLUSIONS Insulin resistance is associated with the aggressiveness of PDAC.
Collapse
Affiliation(s)
- Erica Dugnani
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Gianpaolo Balzano
- Pancreatic Surgery Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Valentina Pasquale
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Marina Scavini
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Francesca Aleotti
- Pancreatic Surgery Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Daniela Liberati
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Gaetano Di Terlizzi
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Alessandra Gandolfi
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Giovanna Petrella
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Michele Reni
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Claudio Doglioni
- Department of Pathology, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Emanuele Bosi
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Massimo Falconi
- Pancreatic Surgery Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
38
|
Navarrete-Muñoz EM, Wark PA, Romaguera D, Bhoo-Pathy N, Michaud D, Molina-Montes E, Tjønneland A, Olsen A, Overvad K, Boutron-Ruault MC, Clavel-Chapelon F, Fagherazzi G, Katzke VA, Kühn T, Steffen A, Trichopoulou A, Klinaki E, Papatesta EM, Masala G, Krogh V, Tumino R, Naccarati A, Mattiello A, Peeters PH, Rylander C, Parr CL, Skeie G, Weiderpass E, Quirós JR, Duell EJ, Dorronsoro M, Huerta JM, Ardanaz E, Wareham N, Khaw KT, Travis RC, Key T, Stepien M, Freisling H, Riboli E, Bueno-de-Mesquita HB. Sweet-beverage consumption and risk of pancreatic cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC). Am J Clin Nutr 2016; 104:760-8. [PMID: 27510540 PMCID: PMC6241849 DOI: 10.3945/ajcn.116.130963] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/23/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The consumption of sweet beverages has been associated with greater risk of type 2 diabetes and obesity, which may be involved in the development of pancreatic cancer. Therefore, it has been hypothesized that sweet beverages may increase pancreatic cancer risk as well. OBJECTIVE We examined the association between sweet-beverage consumption (including total, sugar-sweetened, and artificially sweetened soft drink and juice and nectar consumption) and pancreatic cancer risk. DESIGN The study was conducted within the European Prospective Investigation into Cancer and Nutrition cohort. A total of 477,199 participants (70.2% women) with a mean age of 51 y at baseline were included, and 865 exocrine pancreatic cancers were diagnosed after a median follow-up of 11.60 y (IQR: 10.10-12.60 y). Sweet-beverage consumption was assessed with the use of validated dietary questionnaires at baseline. HRs and 95% CIs were obtained with the use of multivariable Cox regression models that were stratified by age, sex, and center and adjusted for educational level, physical activity, smoking status, and alcohol consumption. Associations with total soft-drink consumption were adjusted for juice and nectar consumption and vice versa. RESULTS Total soft-drink consumption (HR per 100 g/d: 1.03; 95% CI: 0.99, 1.07), sugar-sweetened soft-drink consumption (HR per 100 g/d: 1.02; 95% CI: 0.97, 1.08), and artificially sweetened soft-drink consumption (HR per 100 g/d: 1.04; 95% CI: 0.98, 1.10) were not associated with pancreatic cancer risk. Juice and nectar consumption was inversely associated with pancreatic cancer risk (HR per 100 g/d: 0.91; 95% CI: 0.84, 0.99); this association remained statistically significant after adjustment for body size, type 2 diabetes, and energy intake. CONCLUSIONS Soft-drink consumption does not seem to be associated with pancreatic cancer risk. Juice and nectar consumption might be associated with a modest decreased pancreatic cancer risk. Additional studies with specific information on juice and nectar subtypes are warranted to clarify these results.
Collapse
Affiliation(s)
- Eva M Navarrete-Muñoz
- Department of Public Health, Faculty of Medicine, Miguel Hernández University, Alicante, Spain; The Spanish Biomedical Research Centre in Epidemology and Public Health (CIBERESP), Health Institute Carlos III, Madrid, Spain
| | - Petra A Wark
- Global eHealth Unit, Department of Primary Care and Public Health
| | - Dora Romaguera
- Department of Epidemiology and Biostatistics, and The Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Health Institute Carlos III, Madrid, Spain; Medical Research Institute of Palma, University Hospital Son Espases, Palma de Mallorca, Spain;
| | - Nirmala Bhoo-Pathy
- Julius Centre University of Malaya, Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Dominique Michaud
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA
| | - Esther Molina-Montes
- The Spanish Biomedical Research Centre in Epidemology and Public Health (CIBERESP), Health Institute Carlos III, Madrid, Spain; Andalusian School of Public Health. Biomedical Research Institute of Granada; University Hospital of Granada/Granada University, Granada, Spain
| | - Anne Tjønneland
- Diet, Genes and Environment, Danish Cancer Society Research Center, Copenhagen Ø, Denmark
| | - Anja Olsen
- Diet, Genes and Environment, Danish Cancer Society Research Center, Copenhagen Ø, Denmark
| | - Kim Overvad
- Department of Public Health, Section for Epidemiology, Aarhus University, Aarhus C, Denmark
| | - Marie-Christine Boutron-Ruault
- Centre for Research in Epidemiology and Population Health, U1018, Nutrition, Hormones and Women's Health team, National Institute for Health and Medical Research, Villejuif, France; UMRS 1018, Université Paris Sud, Villejuif, France; Institut Gustave Roussy, Villejuif, France
| | - Françoise Clavel-Chapelon
- Centre for Research in Epidemiology and Population Health, U1018, Nutrition, Hormones and Women's Health team, National Institute for Health and Medical Research, Villejuif, France; UMRS 1018, Université Paris Sud, Villejuif, France; Institut Gustave Roussy, Villejuif, France
| | - Guy Fagherazzi
- Centre for Research in Epidemiology and Population Health, U1018, Nutrition, Hormones and Women's Health team, National Institute for Health and Medical Research, Villejuif, France; UMRS 1018, Université Paris Sud, Villejuif, France; Institut Gustave Roussy, Villejuif, France
| | - Verena A Katzke
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Tilman Kühn
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Annika Steffen
- Department of Epidemiology, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
| | - Antonia Trichopoulou
- Hellenic Health Foundation, Athens, Greece; Bureau of Epidemiologic Research, Academy of Athens, Athens, Greece
| | | | | | - Giovanna Masala
- Molecular and Nutritional Epidemiology Unit, Cancer Research and Prevention Institute-ISPO, Florence, Italy
| | - Vittorio Krogh
- Epidemiology and Prevention Unit. Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Unit, "Civic - M.P. Arezzo" Hospital, ASP Ragusa, Italy
| | - Alessio Naccarati
- Human Genetics Foundation,Torino, Molecular and Genetic Epidemiology Unit, Torino, Italy
| | - Amalia Mattiello
- Dipartamento di Medicina Clinica e Chirurgia, Federico II University of Naples, Naples, Italy
| | - Petra H Peeters
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom; Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Netherlands
| | - Charlotta Rylander
- Department of Community Medicine, University of Tromsø-the Arctic University of Norway, Tromsø, Norway
| | - Christine L Parr
- Department of Chronic Diseases, Division of Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Guri Skeie
- Department of Community Medicine, University of Tromsø-the Arctic University of Norway, Tromsø, Norway
| | - Elisabete Weiderpass
- Department of Community Medicine, University of Tromsø-the Arctic University of Norway, Tromsø, Norway; Department of Research, Cancer Registry of Norway, Oslo, Norway; Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden; Samfundet Folkhälsan, Helsinki, Finland
| | | | - Eric J Duell
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Bellvitge Biomedical Research Institute, Catalan Institute of Oncology, Barcelona, Spain
| | - Miren Dorronsoro
- The Spanish Biomedical Research Centre in Epidemology and Public Health (CIBERESP), Health Institute Carlos III, Madrid, Spain; Public Health Direction Biodonostia Basque Regional Health Department, San Sebastian, Spain
| | - José María Huerta
- The Spanish Biomedical Research Centre in Epidemology and Public Health (CIBERESP), Health Institute Carlos III, Madrid, Spain; Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia, Spain
| | - Eva Ardanaz
- The Spanish Biomedical Research Centre in Epidemology and Public Health (CIBERESP), Health Institute Carlos III, Madrid, Spain; Navarre Public Health Institute, Pamplona, Spain
| | | | - Kay-Tee Khaw
- Department of Public Health and Primary Care, and Clinical Gerontology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Ruth C Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Tim Key
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Magdalena Stepien
- Nutrition and Metabolism Section, International Agency for Research on Cancer, Lyon, France
| | - Heinz Freisling
- Department for Determinants of Chronic Diseases, National Institute for Public Health and the Environment, Bilthoven, Netherlands; and
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, and
| | - H Bas Bueno-de-Mesquita
- Department of Epidemiology and Biostatistics, and Julius Centre University of Malaya, Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Department for Determinants of Chronic Diseases, National Institute for Public Health and the Environment, Bilthoven, Netherlands; and Department of Gastroenterology and Hepatology, University Medical Center, Utrecht, Netherlands
| |
Collapse
|
39
|
Hope C, Robertshaw A, Cheung KL, Idris I, English E. Relationship between HbA1c and cancer in people with or without diabetes: a systematic review. Diabet Med 2016; 33:1013-25. [PMID: 26577885 DOI: 10.1111/dme.13031] [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] [Accepted: 11/11/2015] [Indexed: 12/25/2022]
Abstract
AIM To identify the relationship between HbA1c and cancers in people with or without diabetes. BACKGROUND Cancer is a major public health problem, accounting for 8.2 million deaths worldwide in 2012. HbA1c level has been associated with the risk of developing certain cancers, although the existing evidence is conflicting. METHODS EMBASE, MEDLINE, CINAHL and the Cochrane Library were searched. Eligible articles included randomized controlled trials, cohort studies, case-control studies, systematic reviews and meta-analyses. Participants of either sex, with or without Type 1 or 2 diabetes, were included. The studies were assessed using the Scottish Intercollegiate Guidelines Network (SIGN) criteria by two independent assessors. No meta-analysis was performed because of the heterogeneity of results. RESULTS A total of 19 studies from 1006 met the inclusion criteria, of which 14 were cohort studies and five were nested case-control studies. Eight studies investigated outcomes for all cancer sites. Four of these studies reported that higher HbA1c levels were associated with higher incidence and/or mortality risk for all cancers. One study observed a U-shaped relationship between HbA1c and cancer incidence and mortality. Increasing HbA1c levels were associated with increasing risk of developing colorectal, pancreatic, respiratory and female genital tract cancers. No increased risk was observed for breast cancer, gastrointestinal or urological malignancies. CONCLUSION HbA1c appears to be associated with cancer incidence and/or cancer mortality, but further studies are needed to fully understand the complex relationship between HbA1c and cancer.
Collapse
Affiliation(s)
- C Hope
- School of Medicine, University of Nottingham, Royal Derby Hospital, Derby, UK
| | - A Robertshaw
- School of Medicine, University of Nottingham, Royal Derby Hospital, Derby, UK
| | - K L Cheung
- School of Medicine, University of Nottingham, Royal Derby Hospital, Derby, UK
| | - I Idris
- School of Medicine, University of Nottingham, Royal Derby Hospital, Derby, UK
| | - E English
- School of Medicine, University of Nottingham, Royal Derby Hospital, Derby, UK
| |
Collapse
|
40
|
Epidemiology and Inherited Predisposition for Sporadic Pancreatic Adenocarcinoma. Hematol Oncol Clin North Am 2016; 29:619-40. [PMID: 26226901 DOI: 10.1016/j.hoc.2015.04.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Given the changing demographics of Western populations, the numbers of pancreatic cancer cases are projected to increase during the next decade. Diabetes, recent cigarette smoking, and excess body weight are the cancer's most consistent risk factors. The search for common and rare germline variants that influence risk of pancreatic cancer through genome-wide association studies and high-throughput-sequencing-based studies is underway and holds the promise of increasing the knowledge of variants and genes that play a role in inherited susceptibility of this disease. Research reported in this review has advanced the understanding of pancreatic cancer.
Collapse
|
41
|
Biadgo B, Abebe M. Type 2 Diabetes Mellitus and Its Association with the Risk of Pancreatic Carcinogenesis: A Review. THE KOREAN JOURNAL OF GASTROENTEROLOGY = TAEHAN SOHWAGI HAKHOE CHI 2016; 67:168-77. [PMID: 27112242 DOI: 10.4166/kjg.2016.67.4.168] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The prevalence of diabetes mellitus (DM) and associated diseases such as cancers are substantially increasing worldwide. About 80% of the patients with pancreatic cancer have glucose metabolism alterations. This suggests an association between type 2 DM and pancreatic cancer risk and progression. There are hypotheses that show metabolic links between the diseases, due to insulin resistance, hyperglycemia, hyperinsulinemia, low grade chronic inflammation, and alteration in the insulin-insulin-like growth factor axis. The use of diabetes medications can influence the extent of carcinogenesis of the pancreas. This study briefly reviews recent literature on investigation of metabolic link of type 2 DM, risk of carcinogenesis of the pancreas and their association, as well as the current understanding of metabolic pathways implicated in metabolism and cellular growth. The main finding of this review, although there are discrepancies, is that according to most research long-term DM does not raise the risk of pancreatic cancer. The longest duration of DM may reflect hypoinsulinemia due to treatment for hyperglycemia, but recent onset diabetes was associated with increased risk for pancreatic cancer due to hyperinsulinemia and hyperglycemia. In conclusion, the review demonstrates that type 2 DM and the duration of diabetes pose a risk for pancreatic carcinogenesis, and that there is biological link between the diseases.
Collapse
Affiliation(s)
- Belete Biadgo
- Department of Clinical Chemistry, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Molla Abebe
- Department of Clinical Chemistry, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
42
|
Premorbid Obesity and Mortality in Patients With Pancreatic Cancer: A Systematic Review and Meta-analysis. Clin Gastroenterol Hepatol 2016; 14:355-368.e; quiz e32. [PMID: 26460214 PMCID: PMC4919073 DOI: 10.1016/j.cgh.2015.09.036] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Revised: 09/22/2015] [Accepted: 09/30/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Obesity is associated with an increased risk for pancreatic cancer, but it is unclear whether it affects mortality. We performed a systematic review and meta-analysis to assess the association between premorbid obesity and mortality from pancreatic cancer. METHODS We performed a systematic search through January 2015 and identified studies of the association between premorbid obesity (at least 1 year prior to pancreatic cancer diagnosis) and pancreatic cancer-related mortality. We estimated summary adjusted hazard ratio (aHR) with 95% confidence interval (CI), comparing data from obese (body mass index [BMI] ≥30 kg/m(2)) and overweight subjects (BMI, 25.0-29.9 kg/m(2)) with those from individuals with a normal BMI (controls) by using random-effects model. RESULTS We identified 13 studies (including 3 studies that pooled multiple cohorts); 5 studies included only patients with pancreatic cancer, whereas 8 studies evaluated pancreatic cancer-related mortality in cancer-free individuals at inception. In the meta-analysis, we observed increase in pancreatic cancer-related mortality among overweight (aHR, 1.06; 95% CI, 1.02-1.11; I(2) = 0) and obese individuals (aHR, 1.31; 95% CI, 1.20-1.42; I(2) = 43%), compared with controls; the association remained when we analyzed data from only subjects with pancreatic cancer. Each 1 kg/m(2) increase in BMI was associated with 10% increase in mortality (aHR, 1.10; 95% CI, 1.05-1.15) with minimal heterogeneity (I(2) = 0). In the subgroup analysis, obesity was associated with increased mortality in Western populations (11 studies; aHR, 1.32; 95% CI, 1.22-1.42) but not in Asia-Pacific populations (2 studies; aHR, 0.98; 95% CI, 0.76-1.27). CONCLUSIONS In a systematic review and meta-analysis, we associated increasing level of obesity with increased mortality in patients with pancreatic cancer in Western but not Asia-Pacific populations. Strategies to reduce obesity-induced metabolic abnormalities might be developed to treat patients with pancreatic cancer.
Collapse
|
43
|
Lu Y, García Rodríguez LA, Malgerud L, González-Pérez A, Martín-Pérez M, Lagergren J, Bexelius TS. New-onset type 2 diabetes, elevated HbA1c, anti-diabetic medications, and risk of pancreatic cancer. Br J Cancer 2015; 113:1607-14. [PMID: 26575601 PMCID: PMC4705881 DOI: 10.1038/bjc.2015.353] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/04/2015] [Accepted: 09/14/2015] [Indexed: 02/06/2023] Open
Abstract
Background: Associations between type 2 diabetes, anti-diabetic medications and pancreatic cancer are controversial. This study aims to clarify such associations with new-onset type 2 diabetes and repeated measurements of glycated haemoglobin (HbA1c) levels. Methods: A nested case–control study was initiated from the Health Improvement Network (THIN) in UK from 1996 to 2010. Information of pancreatic cancer cases was retrieved electronically from the medical records and manually validated. Control subjects were randomly selected and frequency-matched to the cases on sex, age, and calendar years. Multivariable unconditional logistic regression was performed to estimate odds ratios (OR) and 95% confidence intervals (CI), and adjusted for potential confounders. Results: Among 1 574 768 person-years of follow-up, 529 pancreatic cancer cases and 5000 controls were identified. Type 2 diabetes, or changed HbA1c levels (rather than HbA1c levels at diabetes diagnosis) in diabetes patients (⩾4 mmol mol−1 compared with <0 mmol mol−1) were followed by an increased OR of pancreatic cancer (OR, 2.16, 95% CI 1.72–2.72 and OR, 5.06, 95% CI 1.52–16.87, respectively). Among the anti-diabetic medications in diabetes patients, the OR for insulin users was 25.57 (95% CI 11.55–56.60), sulphonylureas 2.22 (95% CI 1.13, 4.40), and metformin users 1.46 (95% CI 0.85–2.52), compared with no use of any anti-diabetic medications. Conclusions: New-onset type 2 diabetes and, particularly, diabetes with rising HbA1c seem to be independent risk factors for pancreatic cancer. The relation between different anti-diabetic medications and pancreatic cancer seems to vary in strength, with the highest risk among users of insulin.
Collapse
Affiliation(s)
- Yunxia Lu
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 76, Sweden.,Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
| | | | - Linnéa Malgerud
- Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm 171 77, Sweden
| | | | - Mar Martín-Pérez
- Centro Español de Investigación Farmacoepidemiológica, Madrid 28004, Spain
| | - Jesper Lagergren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 76, Sweden.,Division of Cancer Studies, King's College London, London WC2R 2LS, UK
| | - Tomas S Bexelius
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 76, Sweden.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm 171 77, Sweden
| |
Collapse
|
44
|
Stolzenberg-Solomon RZ, Newton CC, Silverman DT, Pollak M, Nogueira LM, Weinstein SJ, Albanes D, Männistö S, Jacobs EJ. Circulating Leptin and Risk of Pancreatic Cancer: A Pooled Analysis From 3 Cohorts. Am J Epidemiol 2015; 182:187-97. [PMID: 26085045 PMCID: PMC4517697 DOI: 10.1093/aje/kwv041] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 02/06/2015] [Indexed: 12/12/2022] Open
Abstract
Adiposity is associated with pancreatic cancer; however, the underlying mechanism(s) is uncertain. Leptin is an adipokine involved in metabolic regulation, and obese individuals have higher concentrations. We conducted a pooled, nested case-control study of cohort participants from the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial, the Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study, and the Cancer Prevention Study II Nutrition Cohort to investigate whether prediagnostic serum leptin was associated with pancreatic cancer. A total of 731 pancreatic adenocarcinoma cases that occurred between 1986 and 2010 were included (maximum follow-up, 23 years). Incidence density-selected controls (n = 909) were matched to cases by cohort, age, sex, race, and blood draw date. Conditional logistic regression was used to calculate odds ratios and 95% confidence intervals. Sex-specific quintiles were based on the distribution of the controls. Overall, serum leptin was not associated with pancreatic cancer (quintile 5 vs. quintile 1: odds ratio = 1.13, 95% confidence interval: 0.75, 1.71; Ptrend = 0.38). There was a significant interaction by follow-up time (P = 0.003), such that elevated risk was apparent only during follow-up of more than 10 years after blood draw (quintile 5 vs. quintile 1: odds ratio = 2.55, 95% confidence interval: 1.23, 5.27; Ptrend = 0.004). Our results support an association between increasing leptin concentration and pancreatic cancer; however, long follow-up is necessary to observe the relationship. Subclinical disease may explain the lack of association during early follow-up.
Collapse
Affiliation(s)
- Rachael Z. Stolzenberg-Solomon
- Correspondence to Dr. Rachael Z. Stolzenberg-Solomon, 9609 Medical Center Drive, Room 6E420, Rockville, MD 20850 (e-mail: )
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Sadr-Azodi O, Gudbjörnsdottir S, Ljung R. Pattern of increasing HbA1c levels in patients with diabetes mellitus before clinical detection of pancreatic cancer - a population-based nationwide case-control study. Acta Oncol 2015; 54:986-92. [PMID: 25734801 DOI: 10.3109/0284186x.2015.1006402] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Diabetes mellitus is a risk factor for pancreatic cancer. Impaired insulin resistance might precede the clinical detection of this cancer by several years. METHODS This was a nested case-control population-based study assessing the pattern of glycated hemoglobin (HbA1c) change before clinical detection of pancreatic cancer in a population of individuals with diabetes mellitus. All patients registered in the Swedish National Diabetes Register with a prescription of an anti-diabetic drug between 2005 and 2011 were identified. For each case of pancreatic cancer, 10 controls were randomly selected, matched for age, sex, and factors related to diabetes mellitus. Multivariable conditional logistic regression was used to calculate odds ratios (ORs) and 95% confidence intervals (CIs) for the association between HbA1c and pancreatic cancer. RESULTS In total, 391 cases and 3910 matched controls were identified. The risk of pancreatic cancer was increased more than two-fold in individuals with the highest HbA1c quartile compared with the lowest (OR 1.96, 95% CI 1.40-2.75). The risk of pancreatic cancer remained elevated when comparing the highest HbA1c quartile measured within five years from the clinical detection of pancreatic cancer to the lowest HbA1c quartile (p-value for trend < 0.05). No association was found between HbA1c and pancreatic cancer if HbA1c was measured > 5 years before the clinical detection of pancreatic cancer. CONCLUSIONS The pattern of increasing HbA1c in patients with diabetes mellitus preceded the clinical detection of pancreatic cancer by up to five years. These findings indicate that there is a lead time of several years during which the development of pancreatic cancer might be detectable through screening in patients with diabetes mellitus.
Collapse
Affiliation(s)
- Omid Sadr-Azodi
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Centre for Clinical Research Sörmland, Uppsala University, Eskilstuna, Sweden
| | - Soffia Gudbjörnsdottir
- National Diabetes Register, Registercentrum VGR, Gothenburg, Sweden
- Department of Medicine, Sahlgrenska University Hospital, University of Gothenburg, Sweden
| | - Rickard Ljung
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
46
|
Dietary food groups intake and cooking methods associations with pancreatic cancer: a case-control study. Indian J Gastroenterol 2015; 34:225-32. [PMID: 26063308 DOI: 10.1007/s12664-015-0573-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 05/13/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND The role of dietary habits in the etiology of pancreatic cancer (PC) has not yet been well elucidated. AIM The aim of the present study was to examine the association of the frequency of different food groups' intake and their cooking methods with PC risk based on a well-designed case-control study. METHODS A case-control study including 307 PC patients and 322 controls referred to four tertiary endosonography centers was conducted from January 2011 to January 2014 to compare the frequency intake of different food items and their cooking methods between cases and controls. RESULTS After adjustment for gender, age, body mass index, years of education, diabetes and alcohol history, smoking status, and opium use, a significant direct relationship was observed between PC risk and intake frequency (time/week) of bread (OR = 1.50; 95 % CI 1.05-2.13; p-value 0.024), rice (OR = 2.10; 95 % CI 1.15-3.82; p for trend 0.034), and red meat (OR = 2.25; 95 % CI 1.22-4.14; p for trend 0.033) (time/day), when comparing the highest category of intake frequency with the lowest, while increasing frequency of fish consumption was associated with a lower risk of PC (OR = 0.93; 95 % CI0.59-1.47; p for trend 0.009). Increasing consumption of barbecuing red meat and deep fried vegetables was associated with 67 % and 70 % increased risk of PC (p-value 0.025 and 0.006, respectively). CONCLUSION Our results indicate that increased frequency of intake of bread, rice, and red meat (especially barbecued) and deep fried vegetables can aggregate PC risk, while increased frequency of fish consumption can protect against PC. However, more studies are still needed.
Collapse
|
47
|
Liao WC, Tu YK, Wu MS, Lin JT, Wang HP, Chien KL. Blood glucose concentration and risk of pancreatic cancer: systematic review and dose-response meta-analysis. BMJ 2015; 350:g7371. [PMID: 25556126 PMCID: PMC4282179 DOI: 10.1136/bmj.g7371] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/30/2014] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To evaluate potential linear and non-linear dose-response relations between blood glucose and risk of pancreatic cancer. DESIGN Systematic review and dose-response meta-analysis of prospective observational studies. DATA SOURCES Search of PubMed, Scopus, and related reviews before 30 November 2013 without language restriction. ELIGIBILITY CRITERIA Prospective studies evaluating the association between blood glucose concentration and pancreatic cancer. Retrospective and cross sectional studies excluded to avoid reverse causality. DATA EXTRACTION AND SYNTHESIS Two reviewers independently extracted relevant information and assessed study quality with the Newcastle-Ottawa scale. Random effects dose-response meta-analysis was conducted to assess potential linear and non-linear dose-response relations. RESULTS Nine studies were included for analysis, with a total of 2408 patients with pancreatic cancer. There was a strong linear dose-response association between fasting blood glucose concentration and the rate of pancreatic cancer across the range of prediabetes and diabetes. No non-linear association was detected. The pooled rate ratio of pancreatic cancer per 0.56 mmol/L (10 mg/dL) increase in fasting blood glucose was 1.14 (95% confidence interval 1.06 to 1.22; P<0.001) without significant heterogeneity. Sensitivity analysis excluding blood glucose categories in the range of diabetes showed similar results (pooled rate ratio per 0.56 mmol/L increase in fasting blood glucose was 1.15, 95% confidence interval 1.05 to 1.27; P=0.003), strengthening the association between prediabetes and pancreatic cancer. CONCLUSIONS Every 0.56 mmol/L increase in fasting blood glucose is associated with a 14% increase in the rate of pancreatic cancer. As prediabetes can be improved or even reversed through lifestyle changes, early detection of prediabetes coupled with lifestyle changes could represent a viable strategy to curb the increasing incidence of pancreatic cancer.
Collapse
Affiliation(s)
- Wei-Chih Liao
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, 7 Chang Shan South Road, Taipei 100, Taiwan Institute of Epidemiology and Preventive Medicine, National Taiwan University, 17 Hsu Chow Road, Taipei 100, Taiwan
| | - Yu-Kang Tu
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, 17 Hsu Chow Road, Taipei 100, Taiwan
| | - Ming-Shiang Wu
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, 7 Chang Shan South Road, Taipei 100, Taiwan
| | - Jaw-Town Lin
- School of Medicine, Fu Jen Catholic University, 510 Zhongzheng Road, New Taipei City 242, Taiwan
| | - Hsiu-Po Wang
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, 7 Chang Shan South Road, Taipei 100, Taiwan
| | - Kuo-Liong Chien
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, 7 Chang Shan South Road, Taipei 100, Taiwan Institute of Epidemiology and Preventive Medicine, National Taiwan University, 17 Hsu Chow Road, Taipei 100, Taiwan
| |
Collapse
|
48
|
|
49
|
Campa D, Mergarten B, De Vivo I, Boutron-Ruault MC, Racine A, Severi G, Nieters A, Katzke VA, Trichopoulou A, Yiannakouris N, Trichopoulos D, Boeing H, Quirós JR, Duell EJ, Molina-Montes E, Huerta JM, Ardanaz E, Dorronsoro M, Khaw KT, Wareham N, Travis RC, Palli D, Pala V, Tumino R, Naccarati A, Panico S, Vineis P, Riboli E, Siddiq A, Bueno-de-Mesquita HB, Peeters PH, Nilsson PM, Sund M, Ye W, Lund E, Jareid M, Weiderpass E, Duarte-Salles T, Kong SY, Stepien M, Canzian F, Kaaks R. Leukocyte telomere length in relation to pancreatic cancer risk: a prospective study. Cancer Epidemiol Biomarkers Prev 2014; 23:2447-54. [PMID: 25103821 DOI: 10.1158/1055-9965.epi-14-0247] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Several studies have examined leukocyte telomere length (LTL) as a possible predictor for cancer at various organ sites. The hypothesis originally motivating many of these studies was that shorter telomeres would be associated with an increase in cancer risk; the results of epidemiologic studies have been inconsistent, however, and suggested positive, negative, or null associations. Two studies have addressed the association of LTL in relation to pancreatic cancer risk and the results are contrasting. METHODS We measured LTL in a prospective study of 331 pancreatic cancer cases and 331 controls in the context of the European Prospective Investigation into Cancer and Nutrition (EPIC). RESULTS We observed that the mean LTL was higher in cases (0.59 ± 0.20) than in controls (0.57 ± 0.17), although this difference was not statistically significant (P = 0.07), and a basic logistic regression model showed no association of LTL with pancreas cancer risk. When adjusting for levels of HbA1c and C-peptide, however, there was a weakly positive association between longer LTL and pancreatic cancer risk [OR, 1.13; 95% confidence interval (CI), 1.01-1.27]. Additional analyses by cubic spline regression suggested a possible nonlinear relationship between LTL and pancreatic cancer risk (P = 0.022), with a statistically nonsignificant increase in risk at very low LTL, as well as a significant increase at high LTL. CONCLUSION Taken together, the results from our study do not support LTL as a uniform and strong predictor of pancreatic cancer. IMPACT The results of this article can provide insights into telomere dynamics and highlight the complex relationship between LTL and pancreatic cancer risk.
Collapse
Affiliation(s)
- Daniele Campa
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Björn Mergarten
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Immaculata De Vivo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. Program in Genetic Epidemiology and Statistical Genetics, Harvard School of Public Health, Boston, Massachusetts
| | - Marie-Christine Boutron-Ruault
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre for research in Epidemiology and Population Health (CESP), U1018, Nutrition, Hormones, and Women's Health team, Villejuif, France. Univ Paris Sud, UMRS 1018, Villejuif, France. IGR, Villejuif, France
| | - Antoine Racine
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre for research in Epidemiology and Population Health (CESP), U1018, Nutrition, Hormones, and Women's Health team, Villejuif, France. Univ Paris Sud, UMRS 1018, Villejuif, France. IGR, Villejuif, France
| | | | - Alexandra Nieters
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| | - Verena A Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Antonia Trichopoulou
- Hellenic Health Foundation, Athens, Greece. Bureau of Epidemiologic Research, Academy of Athens, Athens, Greece
| | - Nikos Yiannakouris
- Hellenic Health Foundation, Athens, Greece. Harokopio University of Athens, Greece
| | - Dimitrios Trichopoulos
- Hellenic Health Foundation, Athens, Greece. Bureau of Epidemiologic Research, Academy of Athens, Athens, Greece. Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | | | - Eric J Duell
- Unit of Nutrition, Environment, and Cancer, Cancer Epidemiology Research Program, Bellvitge Biomedical Research Institute (IDIBELL), Catalan Institute of Oncology (ICO), Barcelona, Spain
| | - Esther Molina-Montes
- Escuela Andaluza de Salud Pública, Instituto de Investigación Biosanitaria de Granada (Granada.ibs), Granada, Spain. CIBER Epidemiology and Public Health CIBERESP, Madrid, Spain
| | - José María Huerta
- CIBER Epidemiology and Public Health CIBERESP, Madrid, Spain. Department of Epidemiology, Murcia Regional Health Council, Murcia, Spain
| | - Eva Ardanaz
- CIBER Epidemiology and Public Health CIBERESP, Madrid, Spain. Navarre Public Health Institute, Pamplona, Spain
| | - Miren Dorronsoro
- Public Health Direction and Biodonostia-Ciberesp Basque Regional Health Department, San Sebastian, Spain
| | - Kay-Tee Khaw
- University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Nicholas Wareham
- University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Ruth C Travis
- Cancer Epidemiology Unit, University of Oxford, Oxford, United Kingdom
| | - Domenico Palli
- Molecular and Nutritional Epidemiology Unit, Cancer Research and Prevention Institute, ISPO, Florence, Italy
| | - Valeria Pala
- Epidemiology and Prevention Unit Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Unit, "Civic - M.P. Arezzo" Hospital, ASP Ragusa, Ragusa, Italy
| | | | - Salvatore Panico
- Dipartimento Di Medicina Clinica e Chirurgia Federico II University, Naples, Italy
| | - Paolo Vineis
- Division of Epidemiology, Public Health and Primary Care, Imperial College, London, United Kingdom
| | - Elio Riboli
- Division of Epidemiology, Public Health and Primary Care, Imperial College, London, United Kingdom
| | - Afshan Siddiq
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London, United Kingdom
| | - H B Bueno-de-Mesquita
- National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands. Department of Gastroenterology and Hepatology, University Medical Centre, Utrecht, the Netherlands. The School of Public Health, Imperial College London, London, United Kingdom
| | - Petra H Peeters
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center, Utrecht, the Netherlands
| | - Peter M Nilsson
- Lund University, Department of Clinical Sciences, Skåne University Hospital, Malmö Sweden
| | - Malin Sund
- Department of Surgical and Perioperative Sciences, Umeå University, Umeå, Sweden
| | - Weimin Ye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden. The Medical Biobank at Umeå University, Umeå, Sweden
| | - Eiliv Lund
- Department of Community Medicine, Faculty of Health Sciences, University of Tromso, The Arctic University of Norway, Tromsø, Norway
| | - Mie Jareid
- Department of Community Medicine, Faculty of Health Sciences, University of Tromso, The Arctic University of Norway, Tromsø, Norway
| | - Elisabete Weiderpass
- Department of Community Medicine, Faculty of Health Sciences, University of Tromso, The Arctic University of Norway, Tromsø, Norway. Department of Research, Cancer Registry of Norway, Oslo, Norway. Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden. Samfundet Folkhälsan, Helsinki, Finland
| | | | - So Yeon Kong
- International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Magdalena Stepien
- International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
50
|
Abstract
Type 2 diabetes mellitus is an independent risk factor for cancer such as pancreatic, liver, colorectal and breast cancer. In addition, diabetes decreases the risk of prostate cancer. These associations have been found in numerous epidemiological studies, among them several prospective cohorts. However, such studies do not prove causality of the association and cannot exclude inadequate correction for known confounders (e.g. visceral fat) or the influence of unknown confounders. Thus, it is unclear whether the cancer risk is increased by the causes (e.g. the metabolic syndrome), the metabolic consequences (e.g. hyperglycaemia) or the therapy of diabetes; a question which is of major importance for therapeutic guidelines. Identification of the potential mechanisms by which the diabetes or its therapy accelerates or inhibits the development of cancer will help answering the question by providing biological plausibility. This review will summarize the evidence supporting the association of cancer and type 2 diabetes and discuss its potential causes.
Collapse
Affiliation(s)
- Hans-Georg Joost
- German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany German Center for Diabetes Research, Neuherberg, Germany
| |
Collapse
|