1
|
Wang S, Zhang T, Sun Y, Yang D, Cao X. No genetic causality between appendectomy and gastrointestinal cancers: a Mendelian randomization study and meta-analysis in European population. Sci Rep 2024; 14:25959. [PMID: 39472519 PMCID: PMC11522438 DOI: 10.1038/s41598-024-77600-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024] Open
Abstract
The impact of appendectomy on the risk of gastrointestinal cancers remains unknown. We aimed to systematically estimate the causal relationship between appendectomy and gastrointestinal cancers in the European population using two-sample Mendelian randomization (TSMR) study methods and meta-analysis. As part of the discovery cohort analysis, we identified independent genetic variants strongly associated with appendectomy from the UK Biobank (50,105 cases) to serve as instrumental variables (IVs). Summary-level data for gastrointestinal cancers were obtained from the FinnGen study. As the replication cohort, IVs associated with appendectomy were extracted in the FinnGen study (28,601 cases). The data for gastrointestinal cancers were obtained from the UK Biobank. Finally, meta-analyses were conducted to evaluate the combined causal effects of the MR results. We found no causal relationship between appendectomy and gastrointestinal cancers in both the discovery and replication cohorts. Finally, the meta-analysis revealed no causal association between appendectomy and gastrointestinal cancers. Our findings suggest no causal relationship exists between appendectomy and gastrointestinal cancers in the European population. This genetic evidence supports the conclusion from other observational studies that appendectomy does not affect the risk of gastrointestinal cancers in the European population.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Tao Zhang
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuanlin Sun
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dongliang Yang
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xueyuan Cao
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
2
|
Grigorescu RR, Husar-Sburlan IA, Gheorghe C. Pancreatic Cancer: A Review of Risk Factors. Life (Basel) 2024; 14:980. [PMID: 39202722 PMCID: PMC11355429 DOI: 10.3390/life14080980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Pancreatic adenocarcinoma is one of the most lethal types of gastrointestinal cancer despite the latest medical advances. Its incidence has continuously increased in recent years in developed countries. The location of the pancreas can result in the initial symptoms of neoplasia being overlooked, which can lead to a delayed diagnosis and a subsequent reduction in the spectrum of available therapeutic options. The role of modifiable risk factors in pancreatic cancer has been extensively studied in recent years, with smoking and alcohol consumption identified as key contributors. However, the few screening programs that have been developed focus exclusively on genetic factors, without considering the potential impact of modifiable factors on disease occurrence. Thus, fully understanding and detecting the risk factors for pancreatic cancer represents an important step in the prevention and early diagnosis of this type of neoplasia. This review reports the available evidence on different risk factors and identifies the areas that could benefit the most from additional studies.
Collapse
Affiliation(s)
- Raluca Roxana Grigorescu
- Gastroenterology Department, “Sfanta Maria” Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | | | - Cristian Gheorghe
- Center for Digestive Disease and Liver Transplantation, Fundeni Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
3
|
Rosato V, Gómez-Rubio P, Molina-Montes E, Márquez M, Löhr M, O'Rorke M, Michalski CW, Molero X, Farré A, Perea J, Kleeff J, Crnogorac-Jurcevic T, Greenhalf W, Ilzarbe L, Tardón A, Gress T, Barberá VM, Domínguez-Muñoz E, Muñoz-Bellvís L, Balsells J, Costello E, Iglesias M, Kong B, Mora J, O'Driscoll D, Poves I, Scarpa A, Ye W, Hidalgo M, Sharp L, Carrato A, Real FX, La Vecchia C, Malats N. Gallbladder disease and pancreatic cancer risk: a multicentric case-control European study. Eur J Cancer Prev 2021; 30:423-430. [PMID: 34545020 DOI: 10.1097/cej.0000000000000588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND AIMS The overall evidence on the association between gallbladder conditions (GBC: gallstones and cholecystectomy) and pancreatic cancer (PC) is inconsistent. To our knowledge, no previous investigations considered the role of tumour characteristics on this association. Thus, we aimed to assess the association between self-reported GBC and PC risk, by focussing on timing to PC diagnosis and tumour features (stage, location, and resection). METHODS Data derived from a European case-control study conducted between 2009 and 2014 including 1431 PC cases and 1090 controls. We used unconditional logistic regression models to estimate odds ratios (ORs) and corresponding 95% confidence intervals (CIs) adjusted for recognized confounders. RESULTS Overall, 298 (20.8%) cases and 127 (11.6%) controls reported to have had GBC, corresponding to an OR of 1.70 (95% CI 1.33-2.16). The ORs were 4.84 (95% CI 2.96-7.89) for GBC diagnosed <3 years before PC and 1.06 (95% CI 0.79-1.41) for ≥3 years. The risk was slightly higher for stage I/II (OR = 1.71, 95% CI 1.15-2.55) vs. stage III/IV tumours (OR = 1.23, 95% CI 0.87-1.76); for tumours sited in the head of the pancreas (OR = 1.59, 95% CI 1.13-2.24) vs. tumours located at the body/tail (OR = 1.02, 95% CI 0.62-1.68); and for tumours surgically resected (OR = 1.69, 95% CI 1.14-2.51) vs. non-resected tumours (OR = 1.25, 95% CI 0.88-1.78). The corresponding ORs for GBC diagnosed ≥3 years prior PC were close to unity. CONCLUSION Our study supports the association between GBC and PC. Given the time-risk pattern observed, however, this relationship may be non-causal and, partly or largely, due to diagnostic attention and/or reverse causation.
Collapse
Affiliation(s)
- V Rosato
- Unit of Medical Statistics and Biometry, National Cancer Institute, IRCCS Foundation, Milan, Italy
| | - P Gómez-Rubio
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid
- CIBERONC, Spain
| | - E Molina-Montes
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid
- CIBERONC, Spain
| | - M Márquez
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid
- CIBERONC, Spain
| | - M Löhr
- Gastrocentrum, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - M O'Rorke
- Centre for Public Health, Belfast, Queen's University Belfast, Belfast, UK
| | - C W Michalski
- Department of Surgery, Technical University of Munich, Munich
- Department of Visceral, Vascular and Endocrine Surgery, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - X Molero
- Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute (VHIR), Barcelona, Universitat Autònoma de Barcelona, Barcelona
- CIBEREHD
| | - A Farré
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, Barcelona
| | - J Perea
- Department of Surgery, University Hospital 12 de Octubre
- Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - J Kleeff
- Department of Surgery, Technical University of Munich, Munich
- Department of Visceral, Vascular and Endocrine Surgery, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - T Crnogorac-Jurcevic
- Barts Cancer Institute, Centre for Molecular Oncology, Queen Mary University of London, John Vane Science Centre, London
| | - W Greenhalf
- Department of Molecular and Clinical Cancer Medicine, The Royal Liverpool University Hospital, Liverpool, UK
| | - L Ilzarbe
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid
- Hospital del Mar-Parc de Salut Mar, Barcelona
| | - A Tardón
- Department of Medicine, Instituto Universitario de Oncología del Principado de Asturias, Oviedo
- CIBERESP, Spain
| | - T Gress
- Department of Gastroenterology, University Hospital of Giessen and Marburg, Marburg, Germany
| | - V M Barberá
- Molecular Genetics Laboratory, General University Hospital of Elche
| | - E Domínguez-Muñoz
- Department of Gastroenterology, University Clinical Hospital of Santiago de Compostela
| | - L Muñoz-Bellvís
- General and Digestive Surgery Department, Salamanca University Hospital, Elche, Santiago de Compostela, and Salamanca, Spain
| | - J Balsells
- Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute (VHIR), Barcelona, Universitat Autònoma de Barcelona, Barcelona
- CIBEREHD
| | - E Costello
- Department of Molecular and Clinical Cancer Medicine, The Royal Liverpool University Hospital, Liverpool, UK
| | - M Iglesias
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid
- Hospital del Mar-Parc de Salut Mar, Barcelona
| | - Bo Kong
- Department of Surgery, Technical University of Munich, Munich
| | - J Mora
- Department of Gastroenterology, Hospital de la Santa Creu i Sant Pau, Barcelona
| | - D O'Driscoll
- National Cancer Registry Ireland and HRB Clinical Research Facility, University College Cork, Cork, Ireland
| | - I Poves
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid
- Hospital del Mar-Parc de Salut Mar, Barcelona
| | - A Scarpa
- ARC-Net centre for Applied Research on Cancer and Department of Pathology and Diagnostics, University and Hospital trust of Verona, Verona, Italy
| | - W Ye
- Gastrocentrum, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - M Hidalgo
- Madrid-Norte-Sanchinarro Hospital, Madrid, Spain
| | - L Sharp
- National Cancer Registry Ireland and HRB Clinical Research Facility, University College Cork, Cork, Ireland
- Newcastle University, Institute of Health & Society, Newcastle, UK
| | - A Carrato
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid
- Department of Oncology, Ramón y Cajal University Hospital, IRYCIS, Alcala University
| | - F X Real
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid
- Epithelial Carcinogenesis Group
- Spanish National Cancer Research Centre (CNIO), Madrid
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - C La Vecchia
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - N Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid
- CIBERONC, Spain
| |
Collapse
|
4
|
Monroy-Iglesias MJ, Dolly S, Sarker D, Thillai K, Van Hemelrijck M, Santaolalla A. Pancreatic Cancer Exposome Profile to Aid Early Detection and Inform Prevention Strategies. J Clin Med 2021; 10:1665. [PMID: 33924591 PMCID: PMC8069449 DOI: 10.3390/jcm10081665] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/19/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PCa) is associated with a poor prognosis and high mortality rate. The causes of PCa are not fully elucidated yet, although certain exposome factors have been identified. The exposome is defined as the sum of all environmental factors influencing the occurrence of a disease during a life span. The development of an exposome approach for PCa has the potential to discover new disease-associated factors to better understand the carcinogenesis of PCa and help with early detection strategies. Our systematic review of the literature identified several exposome factors that have been associated with PCa alone and in combination with other exposures. A potential inflammatory signature has been observed among the interaction of several exposures (i.e., smoking, alcohol consumption, diabetes mellitus, obesity, and inflammatory markers) that further increases the incidence and progression of PCa. A large number of exposures have been identified such as genetic, hormonal, microorganism infections and immune responses that warrant further investigation. Future early detection strategies should utilize this information to assess individuals' risk for PCa.
Collapse
Affiliation(s)
- Maria J. Monroy-Iglesias
- Translational Oncology & Urology Research (TOUR), School of Cancer and Pharmaceutical Sciences, King’s College London, London SE1 9RT, UK; (M.J.M.-I.); (M.V.H.)
| | - Saoirse Dolly
- Department of Medical Oncology, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 9RT, UK; (S.D.); (D.S.); (K.T.)
| | - Debashis Sarker
- Department of Medical Oncology, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 9RT, UK; (S.D.); (D.S.); (K.T.)
- School of Cancer and Pharmaceutical Sciences, King’s College London, London SE1 9RT, UK
| | - Kiruthikah Thillai
- Department of Medical Oncology, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 9RT, UK; (S.D.); (D.S.); (K.T.)
| | - Mieke Van Hemelrijck
- Translational Oncology & Urology Research (TOUR), School of Cancer and Pharmaceutical Sciences, King’s College London, London SE1 9RT, UK; (M.J.M.-I.); (M.V.H.)
| | - Aida Santaolalla
- Translational Oncology & Urology Research (TOUR), School of Cancer and Pharmaceutical Sciences, King’s College London, London SE1 9RT, UK; (M.J.M.-I.); (M.V.H.)
| |
Collapse
|
5
|
The Epidemiology of Pancreatic Cancer and the Association With Acetylsalicylic Acid in the United States: A Population-Based Study. Pancreas 2020; 49:1207-1212. [PMID: 32898007 DOI: 10.1097/mpa.0000000000001659] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Pancreatic cancer (PaC) is the third leading cause of cancer-related death in the United States. Multiple studies have investigated the epidemiology and the association between PaC and acetylsalicylic acid (ASA) use, revealing mixed results. Using a large database, we sought to investigate the epidemiology of PaC. METHODS Using a commercial database (Explorys Inc, Cleveland, Ohio), which includes electronic health record data from 26 major integrated US health care systems, all patients 18 years and older diagnosed with PaC for 5 years were identified based on Systematized Nomenclature Of Medicine-Clinical Terms. We determined the prevalence of PaC and the potential associated factors using univariable and multivariable analysis. RESULTS Of the 32,970,850 individuals, we identified 30,250 individuals with PaC with an overall prevalence of 0.08%. Individuals with PaC were more likely to be males, seniors (age, >65 years), and White, compared with patients without PaC. In subgroup analysis of chronic pancreatitis, individuals on ASA, individuals >65 years, White, and White males were less likely to have PaC, and non-White females were more likely to have PaC. CONCLUSIONS This is the largest population-based study evaluating the epidemiology of PaC. We observed a protective association of ASA on a subgroup of patients with PaC and chronic pancreatitis.
Collapse
|
6
|
Rosato V, Negri E, Bosetti C, Malats N, Gomez-Rubio P, Consortium P, Maisonneuve P, Miller AB, Bueno-de-Mesquita HB, Baghurst PA, Zatonski W, Petersen GM, Scelo G, Holcatova I, Fabianova E, Serraino D, Olson SH, Vioque J, Lagiou P, Duell EJ, Boffetta P, La Vecchia C. Gallbladder disease, cholecystectomy, and pancreatic cancer risk in the International Pancreatic Cancer Case-Control Consortium (PanC4). Eur J Cancer Prev 2020; 29:408-415. [PMID: 32740166 DOI: 10.1097/cej.0000000000000572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The association among gallbladder disease, cholecystectomy, and pancreatic cancer is unclear. Moreover, time interval between gallbladder disease or cholecystectomy and pancreatic cancer diagnosis is not considered in most previous studies. AIM To quantify the association among gallbladder disease, cholecystectomy, and pancreatic cancer, considering time since first diagnosis of gallbladder disease or cholecystectomy. METHODS We used data from nine case-control studies within the Pancreatic Cancer Case-Control Consortium, including 5760 cases of adenocarcinoma of the exocrine pancreas and 8437 controls. We estimated pooled odds ratios and the corresponding 95% confidence intervals by estimating study-specific odds ratios through multivariable unconditional logistic regression models, and then pooling the obtained estimates using fixed-effects models. RESULTS Compared with patients with no history of gallbladder disease, the pooled odds ratio of pancreatic cancer was 1.69 (95% confidence interval, 1.51-1.88) for patients reporting a history of gallbladder disease. The odds ratio was 4.90 (95% confidence interval, 3.45-6.97) for gallbladder disease diagnosed <2 years before pancreatic cancer diagnosis and 1.11 (95% confidence interval, 0.96-1.29) when ≥2 years elapsed. The pooled odds ratio was 1.64 (95% confidence interval, 1.43-1.89) for patients who underwent cholecystectomy, as compared to those without cholecystectomy. The odds ratio was 7.00 (95% confidence interval, 4.13-11.86) for a surgery <2 years before pancreatic cancer diagnosis and 1.28 (95% confidence interval, 1.08-1.53) for a surgery ≥2 years before. CONCLUSIONS There appears to be no long-term effect of gallbladder disease on pancreatic cancer risk, and at most a modest one for cholecystectomy. The strong short-term association can be explained by diagnostic bias and reverse causation.
Collapse
Affiliation(s)
- Valentina Rosato
- Unit of Medical Statistics and Biometry, National Cancer Institute, IRCCS Foundation
| | - Eva Negri
- Department of Biomedical and Clinical Sciences, University of Milan
| | - Cristina Bosetti
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO) and CIBERONC, Madrid, Spain
| | - Paulina Gomez-Rubio
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO) and CIBERONC, Madrid, Spain
| | - PanGenEU Consortium
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO) and CIBERONC, Madrid, Spain
| | - Patrick Maisonneuve
- Division of Epidemiology and Biostatistics, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Anthony B Miller
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - H Bas Bueno-de-Mesquita
- National Institute for Public Health and the Environment (RIVM), Bilthoven
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht (UMCU), Utrecht, The Netherlands
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Peter A Baghurst
- Public Health, Women's and Children's Hospital, Adelaide, South Australia, Australia
| | - Witold Zatonski
- Health Promotion Foundation, Nadarzyn
- Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Gloria M Petersen
- Department of Health Sciences Research, Medicine and Medical Genetics, Mayo Clinic, Rochester, New York, USA
| | - Ghislaine Scelo
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Ivana Holcatova
- Institute of Hygiene and Epidemiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Eleonora Fabianova
- Regional Authority of Public Health, Banská Bystrica, Slovak Republic
- Faculty of Health, Catholic University, Ružomberok, Slovak Republic
| | - Diego Serraino
- Cancer Epidemiology Unit, National Cancer Institute Centro di Riferimento Oncologico, IRCCS, Aviano, Italy
| | - Sara H Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jesús Vioque
- Institute for Health and Biomedical Research ISABIAL-UMH, Alicante
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Pagona Lagiou
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, University of Athens, Athens, Greece
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Eric J Duell
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Paolo Boffetta
- The Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York, USA
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
7
|
Fereidouni M, Ferns GA, Bahrami A. Current status and perspectives regarding the association between allergic disorders and cancer. IUBMB Life 2020; 72:1322-1339. [PMID: 32458542 DOI: 10.1002/iub.2285] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/05/2020] [Accepted: 03/19/2020] [Indexed: 12/18/2022]
Abstract
While activation of immune system may lead to a lower risk of some diseases, it has been shown that a history of atopic allergic disorders such as asthma, hay fever, eczema, and food allergies could be related to several types of cancer. However, the evidence is not entirely conclusive. Two proposals suggest a possible mechanism for the association between allergic disorders and cancers: immune surveillance and the antigenic stimulation. The association of allergy and cancer may vary by cancer site and the type of exposure. The aim of current review was to summarize the current knowledge of the association between allergic diseases and the risk of cancers with particular emphasis on case-controls and cohort studies to estimate the cancer risk associated with allergy.
Collapse
Affiliation(s)
- Mohammad Fereidouni
- Department of Immunology, Medical school Birjand University of Medical Sciences, Birjand, Iran.,Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Brighton, UK
| | - Afsane Bahrami
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
8
|
Bagni K, Chen IM, Johansen AZ, Dehlendorff C, Jensen BV, Hansen CP, Preus Hasselby J, Holländer NH, Nissen M, Bjerregaard JK, Pfeiffer P, Yilmaz MK, Rasmussen LS, Nielsen SE, Johansen JS. Prognostic impact of Charlson's Age-Comorbidity Index and other risk factors in patients with pancreatic cancer. Eur J Cancer Care (Engl) 2020; 29:e13219. [PMID: 31908093 DOI: 10.1111/ecc.13219] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 12/01/2019] [Accepted: 12/11/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Few studies have evaluated the impact of risk factors and comorbidity on overall survival (OS) in patients with pancreatic ductal adenocarcinoma (PDAC). The aim was to investigate the prognostic importance of Charlson's age-comorbidity index (CACI) and other risk factors on prognosis in a clinical real-world cohort of PDAC patients. METHODS A total of 1,159 patients with PDAC who had received at least one cycle of adjuvant or palliative chemotherapy were included from the Danish BIOPAC study. We analysed OS according to CACI, tobacco smoking, alcohol intake, performance status (PS), BMI and diabetes. Hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated for OS using Cox proportional hazards regression. RESULTS At the end of follow-up, 994 (86%) patients had died. The median OS was 298 days for all patients (range 3-3010) and shortest in patients with stage IV. No association with short OS was seen for CACI > 2, diabetes, alcohol abuse, tobacco smoking, hypertension, and high BMI. Multivariate analysis showed that stage (IV vs. I: HR = 9.05, 95% CI 5.17-15.84), PS (2 vs. 0: HR = 3.67, 2.92-4.61) and treatment with angiotensin-converting enzyme inhibitors (yes vs. no: HR = 1.31, 1.06-1.61) were independent negative prognostic factors. CONCLUSIONS We found that CACI, diabetes, tobacco smoking, alcohol abuse, hypertension, and high BMI were not associated with OS in a real-world cohort of patients with PDAC treated with chemotherapy. Only stage and PS were prognostic parameters.
Collapse
Affiliation(s)
- Karin Bagni
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Inna M Chen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Astrid Z Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian Dehlendorff
- Statistics and Pharmacoepidemiology, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Benny V Jensen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Carsten P Hansen
- Department of Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jane Preus Hasselby
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Niels H Holländer
- Department of Oncology at, Zealand University Hospital, Naestved, Denmark
| | - Mette Nissen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Oncology at, Zealand University Hospital, Naestved, Denmark
| | | | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Mette K Yilmaz
- Department of Oncology, Aalborg University Hospital, Aalborg, Denmark
| | | | - Svend E Nielsen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Oncology and Palliative Care, North Zealand University Hospital, Hillerød, Denmark
| | - Julia S Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Rawla P, Sunkara T, Gaduputi V. Epidemiology of Pancreatic Cancer: Global Trends, Etiology and Risk Factors. World J Oncol 2019; 10:10-27. [PMID: 30834048 PMCID: PMC6396775 DOI: 10.14740/wjon1166] [Citation(s) in RCA: 1354] [Impact Index Per Article: 270.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 11/14/2018] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer is the seventh leading cause of cancer-related deaths worldwide. However, its toll is higher in more developed countries. Reasons for vast differences in mortality rates of pancreatic cancer are not completely clear yet, but it may be due to lack of appropriate diagnosis, treatment and cataloging of cancer cases. Because patients seldom exhibit symptoms until an advanced stage of the disease, pancreatic cancer remains one of the most lethal malignant neoplasms that caused 432,242 new deaths in 2018 (GLOBOCAN 2018 estimates). Globally, 458,918 new cases of pancreatic cancer have been reported in 2018, and 355,317 new cases are estimated to occur until 2040. Despite advancements in the detection and management of pancreatic cancer, the 5-year survival rate still stands at 9% only. To date, the causes of pancreatic carcinoma are still insufficiently known, although certain risk factors have been identified, such as tobacco smoking, diabetes mellitus, obesity, dietary factors, alcohol abuse, age, ethnicity, family history and genetic factors, Helicobacter pylori infection, non-O blood group and chronic pancreatitis. In general population, screening of large groups is not considered useful to detect the disease at its early stage, although newer techniques and the screening of tightly targeted groups (especially of those with family history), are being evaluated. Primary prevention is considered of utmost importance. Up-to-date statistics on pancreatic cancer occurrence and outcome along with a better understanding of the etiology and identifying the causative risk factors are essential for the primary prevention of this disease.
Collapse
Affiliation(s)
- Prashanth Rawla
- Department of Internal Medicine, SOVAH Health, Martinsville, VA 24112, USA
| | - Tagore Sunkara
- Department of Gastroenterology and Hepatology, Mercy Medical Center, Des Moines, IA 50314, USA
| | - Vinaya Gaduputi
- Division of Gastroenterology, SBH Health System, Bronx, NY, USA
| |
Collapse
|
10
|
Gomez-Rubio P, Rosato V, Márquez M, Bosetti C, Molina-Montes E, Rava M, Piñero J, Michalski CW, Farré A, Molero X, Löhr M, Ilzarbe L, Perea J, Greenhalf W, O'Rorke M, Tardón A, Gress T, Barberá VM, Crnogorac-Jurcevic T, Muñoz-Bellvís L, Domínguez-Muñoz E, Gutiérrez-Sacristán A, Balsells J, Costello E, Guillén-Ponce C, Huang J, Iglesias M, Kleeff J, Kong B, Mora J, Murray L, O'Driscoll D, Peláez P, Poves I, Lawlor RT, Carrato A, Hidalgo M, Scarpa A, Sharp L, Furlong LI, Real FX, La Vecchia C, Malats N. A systems approach identifies time-dependent associations of multimorbidities with pancreatic cancer risk. Ann Oncol 2017; 28:1618-1624. [PMID: 28383714 DOI: 10.1093/annonc/mdx167] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is usually diagnosed in late adulthood; therefore, many patients suffer or have suffered from other diseases. Identifying disease patterns associated with PDAC risk may enable a better characterization of high-risk patients. METHODS Multimorbidity patterns (MPs) were assessed from 17 self-reported conditions using hierarchical clustering, principal component, and factor analyses in 1705 PDAC cases and 1084 controls from a European population. Their association with PDAC was evaluated using adjusted logistic regression models. Time since diagnosis of morbidities to PDAC diagnosis/recruitment was stratified into recent (<3 years) and long term (≥3 years). The MPs and PDAC genetic networks were explored with DisGeNET bioinformatics-tool which focuses on gene-diseases associations available in curated databases. RESULTS Three MPs were observed: gastric (heartburn, acid regurgitation, Helicobacter pylori infection, and ulcer), metabolic syndrome (obesity, type-2 diabetes, hypercholesterolemia, and hypertension), and atopic (nasal allergies, skin allergies, and asthma). Strong associations with PDAC were observed for ≥2 recently diagnosed gastric conditions [odds ratio (OR), 6.13; 95% confidence interval CI 3.01-12.5)] and for ≥3 recently diagnosed metabolic syndrome conditions (OR, 1.61; 95% CI 1.11-2.35). Atopic conditions were negatively associated with PDAC (high adherence score OR for tertile III, 0.45; 95% CI, 0.36-0.55). Combining type-2 diabetes with gastric MP resulted in higher PDAC risk for recent (OR, 7.89; 95% CI 3.9-16.1) and long-term diagnosed conditions (OR, 1.86; 95% CI 1.29-2.67). A common genetic basis between MPs and PDAC was observed in the bioinformatics analysis. CONCLUSIONS Specific multimorbidities aggregate and associate with PDAC in a time-dependent manner. A better characterization of a high-risk population for PDAC may help in the early diagnosis of this cancer. The common genetic basis between MP and PDAC points to a mechanistic link between these conditions.
Collapse
Affiliation(s)
- P Gomez-Rubio
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, and CIBERONC, Spain
| | - V Rosato
- Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro," Department of Clinical Sciences and Community Health, University of Milan, Milan
- Unit of Medical Statistics, Biometry and Bioinformatics, National Cancer Institute, IRCCS Foundation, Milan
| | - M Márquez
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, and CIBERONC, Spain
| | - C Bosetti
- Department of Epidemiology, Mario Negri Institute for Pharmacological Research-IRCCS, Milan, Italy
| | - E Molina-Montes
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, and CIBERONC, Spain
| | - M Rava
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, and CIBERONC, Spain
| | - J Piñero
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Research Institute (IMIM), Pompeu Fabra Univeristy (UPF), Barcelona, Spain
| | - C W Michalski
- Department of Surgery, Technical University of Munich, Munich
- Department of Surgery, University of Heidelberg, Heidelberg, Germany
| | - A Farré
- Department of Gastroenterology, Santa Creu i Sant Pau Hospital, Barcelona
| | - X Molero
- Exocrine Pancreas Research Unit and Vall d'Hebron Research Institute (VHIR), Vall d'Hebron University Hospital, Barcelona
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona
- Network of Biomedical Research Centres (CIBER), Hepatic and Digestive Diseases and Epidemiology and Public Health, Madrid, Spain
| | - M Löhr
- Gastrocentrum, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - L Ilzarbe
- Department of Gastroenterology, Parc de Salut Mar University Hospital, Barcelona
| | - J Perea
- Department of Surgery, 12 de Octubre University Hospital, Madrid, Spain
| | - W Greenhalf
- Department of Molecular and Clinical Cancer Medicine, The Royal Liverpool University Hospital, Liverpool
| | - M O'Rorke
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - A Tardón
- Network of Biomedical Research Centres (CIBER), Hepatic and Digestive Diseases and Epidemiology and Public Health, Madrid, Spain
- Department of Medicine, University Institute of Oncology of Asturias, Oviedo, Spain
| | - T Gress
- Department of Gastroenterology, University Hospital of Giessen and Marburg, Marburg, Germany
| | - V M Barberá
- Molecular Genetics Laboratory, General University Hospital of Elche, Elche, Spain
| | - T Crnogorac-Jurcevic
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, UK
| | - L Muñoz-Bellvís
- General and Digestive Surgery Department, Salamanca University Hospital, Salamanca
| | - E Domínguez-Muñoz
- Department of Gastroenterology, Clinical University Hospital of Santiago de Compostela, Santiago de Compostela
| | - A Gutiérrez-Sacristán
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Research Institute (IMIM), Pompeu Fabra Univeristy (UPF), Barcelona, Spain
| | - J Balsells
- Exocrine Pancreas Research Unit and Vall d'Hebron Research Institute (VHIR), Vall d'Hebron University Hospital, Barcelona
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona
- Network of Biomedical Research Centres (CIBER), Hepatic and Digestive Diseases and Epidemiology and Public Health, Madrid, Spain
| | - E Costello
- Department of Molecular and Clinical Cancer Medicine, The Royal Liverpool University Hospital, Liverpool
| | - C Guillén-Ponce
- Department of Oncology, Ramón y Cajal Hospital, Madrid, and CIBERONC, Spain
| | - J Huang
- Gastrocentrum, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - M Iglesias
- Department of Gastroenterology, Parc de Salut Mar University Hospital, Barcelona
| | - J Kleeff
- Department of Surgery, Technical University of Munich, Munich
- Department of Molecular and Clinical Cancer Medicine, The Royal Liverpool University Hospital, Liverpool
| | - B Kong
- Department of Surgery, Technical University of Munich, Munich
| | - J Mora
- Department of Gastroenterology, Santa Creu i Sant Pau Hospital, Barcelona
| | - L Murray
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - D O'Driscoll
- Research Programme, National Cancer Registry Ireland
| | - P Peláez
- Department of Surgery, 12 de Octubre University Hospital, Madrid, Spain
| | - I Poves
- Department of Gastroenterology, Parc de Salut Mar University Hospital, Barcelona
| | - R T Lawlor
- ARC-Net Centre for Applied Research on Cancer and Department of Pathology and Diagnostics, University and Hospital trust of Verona, Verona, Italy
| | - A Carrato
- Department of Oncology, Ramón y Cajal Hospital, Madrid, and CIBERONC, Spain
| | - M Hidalgo
- Clara Campal Integrated Oncological Centre, Sanchinarro Hospital, Madrid, Spain
| | - A Scarpa
- ARC-Net Centre for Applied Research on Cancer and Department of Pathology and Diagnostics, University and Hospital trust of Verona, Verona, Italy
| | - L Sharp
- Research Programme, National Cancer Registry Ireland
- Institute of Health & Society, Newcastle University, UK
| | - L I Furlong
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Research Institute (IMIM), Pompeu Fabra Univeristy (UPF), Barcelona, Spain
| | - F X Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre (CNIO), Madrid, and CIBERONC
- Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain
| | - C La Vecchia
- Branch of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro," Department of Clinical Sciences and Community Health, University of Milan, Milan
| | - N Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, and CIBERONC, Spain
| |
Collapse
|
11
|
Barone E, Corrado A, Gemignani F, Landi S. Environmental risk factors for pancreatic cancer: an update. Arch Toxicol 2016; 90:2617-2642. [PMID: 27538405 DOI: 10.1007/s00204-016-1821-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/04/2016] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer (PC) is one of the most aggressive diseases. Only 10 % of all PC cases are thought to be due to genetic factors. Here, we analyzed the most recently published case-control association studies, meta-analyses, and cohort studies with the aim to summarize the main environmental factors that could have a role in PC. Among the most dangerous agents involved in the initiation phase, there are the inhalation of cigarette smoke, and the exposure to mutagenic nitrosamines, organ-chlorinated compounds, heavy metals, and ionizing radiations. Moreover, pancreatitis, high doses of alcohol drinking, the body microbial infections, obesity, diabetes, gallstones and/or cholecystectomy, and the accumulation of asbestos fibers seem to play a crucial role in the progression of the disease. However, some of these agents act both as initiators and promoters in pancreatic acinar cells. Protective agents include dietary flavonoids, marine omega-3, vitamin D, fruit, vegetables, and the habit of regular physical activity. The identification of the factors involved in PC initiation and progression could be of help in establishing novel therapeutic approaches by targeting the molecular signaling pathways responsive to these stimuli. Moreover, the identification of these factors could facilitate the development of strategies for an early diagnosis or measures of risk reduction for high-risk people.
Collapse
Affiliation(s)
- Elisa Barone
- Genetic Unit, Department of Biology, University of Pisa, Via Derna, 1, 56121, Pisa, Italy
| | - Alda Corrado
- Genetic Unit, Department of Biology, University of Pisa, Via Derna, 1, 56121, Pisa, Italy
| | - Federica Gemignani
- Genetic Unit, Department of Biology, University of Pisa, Via Derna, 1, 56121, Pisa, Italy
| | - Stefano Landi
- Genetic Unit, Department of Biology, University of Pisa, Via Derna, 1, 56121, Pisa, Italy.
| |
Collapse
|
12
|
Modifiable and non-modifiable risk factors for pancreatic cancer: A review. Cancer Lett 2016; 381:269-77. [PMID: 27461582 DOI: 10.1016/j.canlet.2016.07.022] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/10/2016] [Accepted: 07/19/2016] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma is associated with a poor prognosis and a high case-fatality rate. The reasons for poor prognosis are low rates of curative resection due to local infiltration and distant metastasis. To increase survival rates of patients with pancreatic cancer, early detection through surveillance and screening is important. However, screening could only be cost-effective in high-risk populations. Identification of significant risk factors therefore assumes significance. Risk factors could be non-modifiable or modifiable. Non-modifiable risk factors include increasing age, familial cancer syndromes, Afro-American race, hereditary and other forms of chronic pancreatitis, diabetes, and non-O blood group. Important modifiable risk factors include smoking, obesity, dietary factors such as non-vegetarian diet, and toxins. Preventive strategies at the population level and an effective screening program targeted at high-risk people may help in prevention and early detection of pancreatic ductal adenocarcinoma.
Collapse
|
13
|
Cotterchio M, Lowcock E, Bider-Canfield Z, Lemire M, Greenwood C, Gallinger S, Hudson T. Association between Variants in Atopy-Related Immunologic Candidate Genes and Pancreatic Cancer Risk. PLoS One 2015; 10:e0125273. [PMID: 25945796 PMCID: PMC4422524 DOI: 10.1371/journal.pone.0125273] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 03/22/2015] [Indexed: 02/06/2023] Open
Abstract
Background Many epidemiology studies report that atopic conditions such as allergies are associated with reduced pancreas cancer risk. The reason for this relationship is not yet understood. This is the first study to comprehensively evaluate the association between variants in atopy-related candidate genes and pancreatic cancer risk. Methods A population-based case-control study of pancreas cancer cases diagnosed during 2011-2012 (via Ontario Cancer Registry), and controls recruited using random digit dialing utilized DNA from 179 cases and 566 controls. Following an exhaustive literature review, SNPs in 180 candidate genes were pre-screened using dbGaP pancreas cancer GWAS data; 147 SNPs in 56 allergy-related immunologic genes were retained and genotyped. Logistic regression was used to estimate age-adjusted odd ratio (AOR) for each variant and false discovery rate was used to adjust Wald p-values for multiple testing. Subsequently, a risk allele score was derived based on statistically significant variants. Results 18 SNPs in 14 candidate genes (CSF2, DENND1B, DPP10, FLG, IL13, IL13RA2, LRP1B, NOD1, NPSR1, ORMDL3, RORA, STAT4, TLR6, TRA) were significantly associated with pancreas cancer risk. After adjustment for multiple comparisons, two LRP1B SNPs remained statistically significant; for example, LRP1B rs1449477 (AA vs. CC: AOR=0.37, 95% CI: 0.22-0.62; p (adjusted)=0.04). Furthermore, the risk allele score was associated with a significant reduction in pancreas cancer risk (p=0.0007). Conclusions Preliminary findings suggest certain atopy-related variants may be associated with pancreas cancer risk. Further studies are needed to replicate this, and to elucidate the biology behind the growing body of epidemiologic evidence suggesting allergies may reduce pancreatic cancer risk.
Collapse
Affiliation(s)
- Michelle Cotterchio
- Prevention and Cancer Control, Cancer Care Ontario, Toronto, ON M5G 2L7, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
- * E-mail:
| | - Elizabeth Lowcock
- Prevention and Cancer Control, Cancer Care Ontario, Toronto, ON M5G 2L7, Canada
| | - Zoe Bider-Canfield
- Prevention and Cancer Control, Cancer Care Ontario, Toronto, ON M5G 2L7, Canada
| | - Mathieu Lemire
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Celia Greenwood
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Steven Gallinger
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Division of General Surgery, Toronto General Hospital, Toronto, ON M5G 2C4, Canada
| | - Thomas Hudson
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
14
|
Wang Y, Zhang FC, Wang YJ. Helicobacter pylori and pancreatic cancer risk: a meta- analysis based on 2,049 cases and 2,861 controls. Asian Pac J Cancer Prev 2015; 15:4449-54. [PMID: 24969867 DOI: 10.7314/apjcp.2014.15.11.4449] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
AIM Helicobacter pylori (H. pylori) have been considered as a risk factor for many cancers. We conducted this meta-analysis to clarify the association between H. pylori infection and the risk of pancreatic cancer. METHODS We searched the Medicine/Pubmed and Embase databases, studies about the association between H. pylori infection and pancreatic cancer published up to Jan.2014 were included. Finally, a total of 9 studies were used for this a meta-analysis. The odds ratios (ORs) and 95% confidence interval (95%CI) of H. pylori infection on pancreatic cancer with respect to control groups were evaluated. Two authors independently assessed the methodological quality and extracted data. This meta-analysis was conducted using software, state (version 12.0) to investigate heterogeneity among individual studies and to summarize the studies. Using the fixed-effects or random-effects model, depending on the absence or presence of significant heterogeneity. Sensitivity analysis was performed to assess the influence of each individual study on the pooled ORs by omitting a single study each time. Publication bias was evaluated by funnel plot, using Egger's and Begg's tests. RESULTS There was no significant association between H. pylori infection and pancreatic cancer risk in the summary ORs,(OR=1.06, 95%CI: 0.74-1.37) through the random-effect method, but heterogeneity among studies was significant (I2=58.9%), so we put the studies into two subgraphs (eastern and western). The results about western (OR=1.14 95%CI:0.89, 1.40) showed heterogeneity among the western countries of I2=6.6%, with no significant association between Hp+ and pancreatic cancer, but the eastern countries (OR=0.62, 95%CI:0.49, 0.76), I2=0, suggested that decreasing pancreas-cancer risk in subjects with Hp+ infection. Simultaneously, 7 studies examined CagA+ strains was (OR=0.84 95%CI:0.63, 1.04), I2=36% with the random-effect method, subgraphs indicated that CagA+ could decrease the risk of pancreatic cancer in the eastern subjects (OR=0.66, 95%CI:0.52-0.80), but the association was not statistically significant in the western subjects (OR=0.95, 95%CI:0.73, 1.16). CONCLUSION Hp+ and CagA+ infection are associated with a decreased risk of pancreatic cancer in eastern populations but have no significant associations in western countries.
Collapse
Affiliation(s)
- Yin Wang
- Postgraduate Training Base of the General Hospital of Jinan Military Command of Liaoning Medical University, Ji Nan, China E-mail :
| | | | | |
Collapse
|
15
|
|
16
|
Klemm C, Dommisch H, Göke F, Kreppel M, Jepsen S, Rolf F, Dommisch K, Perner S, Standop J. Expression profiles for 14-3-3 zeta and CCL20 in pancreatic cancer and chronic pancreatitis. Pathol Res Pract 2014; 210:335-41. [DOI: 10.1016/j.prp.2014.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 11/10/2013] [Accepted: 01/13/2014] [Indexed: 12/17/2022]
|
17
|
Olson SH, Hsu M, Wiemels JL, Bracci PM, Zhou M, Patoka J, Reisacher WR, Wang J, Kurtz RC, Silverman DT, Stolzenberg-Solomon RZ. Serum immunoglobulin e and risk of pancreatic cancer in the prostate, lung, colorectal, and ovarian cancer screening trial. Cancer Epidemiol Biomarkers Prev 2014; 23:1414-20. [PMID: 24718282 DOI: 10.1158/1055-9965.epi-13-1359] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Epidemiologic studies have consistently found that self-reported allergies are associated with reduced risk of pancreatic cancer. Our aim was to prospectively assess the relationship between serum immunoglobulin E (IgE), a marker of allergy, and risk. This nested case-control study within the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO) included subjects enrolled in 1994 to 2001 and followed through 2010. There were 283 cases of pancreatic cancer and 544 controls matched on age, gender, race, and calendar date of blood draw. Using the ImmunoCAP system, we measured total IgE (normal, borderline, elevated), IgE to respiratory allergens, and IgE to food allergens (negative or positive) in serum collected at baseline. Odds ratios (OR) and 95% confidence intervals (CI) were estimated using conditional logistic regression. We assessed interactions with age, gender, smoking, body mass index, and time between randomization and case diagnosis. Overall, there was no association between the IgE measures and risk. We found a statistically significant interaction by baseline age: in those aged ≥65 years, elevated risks were observed for borderline total IgE (OR, 1.43; 95% CI, 0.88-2.32) and elevated total IgE (OR, 1.98; 95% CI, 1.16-3.37) and positive IgE to food allergens (OR, 2.83; 95% CI, 1.29-6.20); among participants <65 years, ORs were <1. Other interactions were not statistically significant. The reduced risk of pancreatic cancer associated with self-reported allergies is not reflected in serum IgE.
Collapse
Affiliation(s)
- Sara H Olson
- Authors' Affiliations: Department of Epidemiology and Biostatistics;
| | - Meier Hsu
- Authors' Affiliations: Department of Epidemiology and Biostatistics
| | - Joseph L Wiemels
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, San Francisco, California
| | - Paige M Bracci
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, San Francisco, California
| | - Mi Zhou
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, San Francisco, California
| | - Joseph Patoka
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, San Francisco, California
| | - William R Reisacher
- Department of Otolaryngology-Head and Neck Surgery, Weill Cornell Medical College, New York, New York
| | - Julie Wang
- Division of Pediatric Allergy and Immunology, Mt. Sinai Medical Center
| | - Robert C Kurtz
- Department of Medicine, Memorial Sloan Kettering Cancer Center
| | | | - Rachael Z Stolzenberg-Solomon
- Branch of Nutritional Epidemiology, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
18
|
Zhang J, Prizment AE, Dhakal IB, Anderson KE. Cholecystectomy, gallstones, tonsillectomy, and pancreatic cancer risk: a population-based case-control study in Minnesota. Br J Cancer 2014; 110:2348-53. [PMID: 24667646 PMCID: PMC4007236 DOI: 10.1038/bjc.2014.154] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 02/27/2014] [Accepted: 02/28/2014] [Indexed: 12/14/2022] Open
Abstract
Background: Associations between medical conditions and pancreatic cancer risk are controversial and are thus evaluated in a study conducted during 1994–1998 in Minnesota. Methods: Cases (n=215) were ascertained from hospitals in the metropolitan area of the Twin Cities and the Mayo Clinic. Controls (n=676) were randomly selected from the general population and frequency matched to cases by age and sex. The history of medical conditions was gathered with a questionnaire during in-person interviews. Odds ratios (OR) and 95% confidence intervals (95% CI) were estimated using unconditional logistic regression. Results: After adjustment for confounders, subjects who had cholecystectomy or gallstones experienced a significantly higher risk of pancreatic cancer than those who did not (OR (95% CI): 2.11 (1.32–3.35) for cholecystectomy and 1.97 (1.23–3.12) for gallstones), whereas opposite results were observed for tonsillectomy (0.67 (0.48–0.94)). Increased risk associated with cholecystectomy was the greatest when it occurred ⩽2 years before the cancer diagnosis (5.93 (2.36–15.7)) but remained statistically significant when that interval was ⩾20 years (2.27 (1.16–4.32)). Conclusions: Cholecystectomy, gallstones, and tonsillectomy were associated with an altered risk of pancreatic cancer. Our study suggests that cholecystectomy increased risk but reverse causality may partially account for high risk associated with recent cholecystectomy.
Collapse
Affiliation(s)
- J Zhang
- 1] Department of Epidemiology, Indiana University, Richard M. Fairbanks School of Public Health at IUPUI, Indianapolis, IN 46202, USA [2] Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN 46202, USA
| | - A E Prizment
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - I B Dhakal
- Department of Biostatistics, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - K E Anderson
- 1] Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN 55455, USA [2] Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
19
|
Cotterchio M, Lowcock E, Hudson TJ, Greenwood C, Gallinger S. Association between allergies and risk of pancreatic cancer. Cancer Epidemiol Biomarkers Prev 2014; 23:469-80. [PMID: 24554712 DOI: 10.1158/1055-9965.epi-13-0965] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Less than 10% of pancreatic cancer cases survive 5 years, yet its etiology is not well understood. Studies suggest allergies are associated with reduced pancreatic cancer risk. Our study collected additional information on allergies (including skin prick test results and differentiation of allergic/nonallergic asthma), and is the first to assess possible confounding by allergy medications. METHODS A population-based case-control study was designed to comprehensively assess the association between allergy and pancreatic cancer risk. Pancreas cancer cases were diagnosed during 2011 to 2012, and identified through the Ontario Cancer Registry (345 cases). Population-based controls were identified using random digit dialing and age/sex frequency matched to cases (1,285 controls). Questionnaires collected lifetime allergy history (type of allergy, age at onset, skin prick testing results), allergy medications, and established pancreas cancer risk factors. Logistic regression was used to estimate odd ratios and test potential confounders, including allergy medications. RESULTS Hay fever was associated with a significant reduction in pancreatic cancer risk [AOR = 0.68; 95% confidence intervals (CI), 0.52-0.89], and reduction was greatest for those whose skin prick test was positive for hay fever allergens. No particular patterns were observed as regards age at onset and duration of allergy. Positive dust/mold allergy skin prick test and animal allergies were associated with a statistically significant reduced pancreatic cancer risk; AOR = 0.49; 95% CI, 0.31-0.78 and AOR = 0.68; 95% CI, 0.46-0.99, respectively. Asthma was not associated with pancreatic cancer risk. CONCLUSIONS/IMPACT These findings support the growing body of evidence that suggests certain allergies are associated with reduced pancreatic cancer risk.
Collapse
Affiliation(s)
- Michelle Cotterchio
- Authors' Affiliations: Prevention and Cancer Control, Cancer Care Ontario; Dalla Lana School of Public Health, University of Toronto; Departments of Medical Biophysics and Molecular Genetics, University of Toronto; Ontario Institute for Cancer Research; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital; and Division of General Surgery, Toronto General Hospital, Toronto, Ontario; and Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | | | | | | | | |
Collapse
|
20
|
Olson SH, Hsu M, Satagopan JM, Maisonneuve P, Silverman DT, Lucenteforte E, Anderson KE, Borgida A, Bracci PM, Bueno-de-Mesquita HB, Cotterchio M, Dai Q, Duell EJ, Fontham EH, Gallinger S, Holly EA, Ji BT, Kurtz RC, La Vecchia C, Lowenfels AB, Luckett B, Ludwig E, Petersen GM, Polesel J, Seminara D, Strayer L, Talamini R. Allergies and risk of pancreatic cancer: a pooled analysis from the Pancreatic Cancer Case-Control Consortium. Am J Epidemiol 2013; 178:691-700. [PMID: 23820785 DOI: 10.1093/aje/kwt052] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In order to quantify the risk of pancreatic cancer associated with history of any allergy and specific allergies, to investigate differences in the association with risk according to age, gender, smoking status, or body mass index, and to study the influence of age at onset, we pooled data from 10 case-control studies. In total, there were 3,567 cases and 9,145 controls. Study-specific odds ratios and 95% confidence intervals were calculated by using unconditional logistic regression adjusted for age, gender, smoking status, and body mass index. Between-study heterogeneity was assessed by using the Cochran Q statistic. Study-specific odds ratios were pooled by using a random-effects model. The odds ratio for any allergy was 0.79 (95% confidence interval (CI): 0.62, 1.00) with heterogeneity among studies (P < 0.001). Heterogeneity was attributable to one study; with that study excluded, the pooled odds ratio was 0.73 (95% CI: 0.64, 0.84) (Pheterogeneity = 0.23). Hay fever (odds ratio = 0.74, 95% CI: 0.56, 0.96) and allergy to animals (odds ratio = 0.62, 95% CI: 0.41, 0.94) were related to lower risk, while there was no statistically significant association with other allergies or asthma. There were no major differences among subgroups defined by age, gender, smoking status, or body mass index. Older age at onset of allergies was slightly more protective than earlier age.
Collapse
Affiliation(s)
- Sara H Olson
- Department of Epidemiology and Biostatistics, 307 East 63rd Street, New York, NY 10065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bosetti C, Lucenteforte E, Bracci PM, Negri E, Neale RE, Risch HA, Olson SH, Gallinger S, Miller AB, Bueno-de-Mesquita HB, Talamini R, Polesel J, Ghadirian P, Baghurst PA, Zatonski W, Fontham E, Holly EA, Gao YT, Yu H, Kurtz RC, Cotterchio M, Maisonneuve P, Zeegers MP, Duell EJ, Boffetta P, La Vecchia C. Ulcer, gastric surgery and pancreatic cancer risk: an analysis from the International Pancreatic Cancer Case-Control Consortium (PanC4). Ann Oncol 2013; 24:2903-10. [PMID: 23970016 DOI: 10.1093/annonc/mdt336] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Peptic ulcer and its treatments have been associated to pancreatic cancer risk, although the evidence is inconsistent. METHODS We pooled 10 case-control studies within the Pancreatic Cancer Case-control Consortium (PanC4), including 4717 pancreatic cancer cases and 9374 controls, and estimated summary odds ratios (OR) using multivariable logistic regression models. RESULTS The OR for pancreatic cancer was 1.10 [95% confidence interval (CI) 0.98-1.23] for history of ulcer (OR = 1.08 for gastric and 0.97 for duodenal ulcer). The association was stronger for a diagnosis within 2 years before cancer diagnosis (OR = 2.43 for peptic, 1.75 for gastric, and 1.98 for duodenal ulcer). The OR was 1.53 (95% CI 1.15-2.03) for history of gastrectomy; however, the excess risk was limited to a gastrectomy within 2 years before cancer diagnosis (OR = 6.18, 95% CI 1.82-20.96), while no significant increased risk was observed for longer time since gastrectomy. No associations were observed for pharmacological treatments for ulcer, such as antacids, H2-receptor antagonists, or proton-pump inhibitors. CONCLUSIONS This uniquely large collaborative study does not support the hypothesis that peptic ulcer and its treatment materially affect pancreatic cancer risk. The increased risk for short-term history of ulcer and gastrectomy suggests that any such association is due to increased cancer surveillance.
Collapse
Affiliation(s)
- C Bosetti
- Department of Epidemiology, IRCCS, Istituto di Ricerche Farmacologiche 'Mario Negri', Milan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Kaur M, Deep G, Jain AK, Raina K, Agarwal C, Wempe MF, Agarwal R. Bitter melon juice activates cellular energy sensor AMP-activated protein kinase causing apoptotic death of human pancreatic carcinoma cells. Carcinogenesis 2013; 34:1585-92. [PMID: 23475945 PMCID: PMC3697895 DOI: 10.1093/carcin/bgt081] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 02/06/2013] [Accepted: 02/21/2013] [Indexed: 12/14/2022] Open
Abstract
Prognosis of pancreatic cancer is extremely poor, suggesting critical needs for additional drugs to improve disease outcome. In this study, we examined efficacy and associated mechanism of a novel agent bitter melon juice (BMJ) against pancreatic carcinoma cells both in culture and nude mice. BMJ anticancer efficacy was analyzed in human pancreatic carcinoma BxPC-3, MiaPaCa-2, AsPC-1 and Capan-2 cells by 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl tetrazolium bromide, cell death enzyme-linked immunosorbent assay and annexin/propidium iodide assays. BMJ effect on apoptosis regulators was assessed by immunoblotting. In vivo BMJ efficacy was evaluated against MiaPaCa-2 tumors in nude mice, and xenograft was analyzed for biomarkers by immunohistochemistry (IHC). Results showed that BMJ (2-5% v/v) decreases cell viability in all four pancreatic carcinoma cell lines by inducing strong apoptotic death. At molecular level, BMJ caused caspases activation, altered expression of Bcl-2 family members and cytochrome-c release into the cytosol. Additionally, BMJ decreased survivin and X-linked inhibitor of apoptosis protein but increased p21, CHOP and phosphorylated mitogen-activated protein kinases (extracellular signal-regulated kinase 1/2 and p38) levels. Importantly, BMJ activated adenosine monophosphate-activated protein kinase (AMPK), a biomarker for cellular energy status, and an AMPK inhibitor (Compound C) reversed BMJ-induced caspase-3 activation suggesting activated AMPK involvement in BMJ-induced apoptosis. In vivo, oral administration of lyophilized BMJ (5mg in 100 µl water/day/mouse) for 6 weeks inhibited MiaPaCa-2 tumor xenograft growth by 60% (P < 0.01) without noticeable toxicity in nude mice. IHC analyses of MiaPaCa-2 xenografts showed that BMJ also inhibits proliferation, induces apoptosis and activates AMPK in vivo. Overall, BMJ exerts strong anticancer efficacy against human pancreatic carcinoma cells, both in vitro and in vivo, suggesting its clinical usefulness.
Collapse
Affiliation(s)
- Manjinder Kaur
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences and
| | - Gagan Deep
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences and
- Department of Pharmaceutical Sciences, University of Colorado Cancer Center, University of Colorado, Aurora, CO 80045, USA
| | - Anil K. Jain
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences and
| | - Komal Raina
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences and
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences and
- Department of Pharmaceutical Sciences, University of Colorado Cancer Center, University of Colorado, Aurora, CO 80045, USA
| | - Michael F. Wempe
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences and
- Department of Pharmaceutical Sciences, University of Colorado Cancer Center, University of Colorado, Aurora, CO 80045, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences and
- Department of Pharmaceutical Sciences, University of Colorado Cancer Center, University of Colorado, Aurora, CO 80045, USA
| |
Collapse
|
23
|
Duell EJ, Lucenteforte E, Olson SH, Bracci PM, Li D, Risch HA, Silverman DT, Ji BT, Gallinger S, Holly EA, Fontham EH, Maisonneuve P, Bueno-de-Mesquita HB, Ghadirian P, Kurtz RC, Ludwig E, Yu H, Lowenfels AB, Seminara D, Petersen GM, La Vecchia C, Boffetta P. Pancreatitis and pancreatic cancer risk: a pooled analysis in the International Pancreatic Cancer Case-Control Consortium (PanC4). Ann Oncol 2012; 23:2964-2970. [PMID: 22767586 PMCID: PMC3477881 DOI: 10.1093/annonc/mds140] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 03/27/2012] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Pancreatitis is a known risk factor for pancreatic cancer; however, an unknown fraction of the disease is thought to be a consequence of tumor-related duct obstruction. PATIENTS AND METHODS A pooled analysis of a history of pancreatitis and risk of pancreatic cancer was carried out considering the time interval between diagnoses and potential modification by covariates. Adjusted pooled odds ratios (ORs) and 95% confidence intervals (CIs) were estimated from 10 case-control studies (5048 cases of ductal pancreatic adenocarcinoma and 10,947 controls) taking part in the International Pancreatic Cancer Case-Control Consortium (PanC4). RESULTS The association between pancreatitis and pancreatic cancer was nearly three-fold at intervals of >2 years between diagnoses (OR: 2.71, 95% CI: 1.96-3.74) and much stronger at intervals of ≤2 years (OR: 13.56, 95% CI: 8.72-21.90) probably reflecting a combination of reverse causation and antecedent misdiagnosis of pancreas cancer as pancreatitis. The younger (<65 years) pancreatic cancer cases showed stronger associations with previous (>2 years) pancreatitis (OR: 3.91, 95% CI: 2.53-6.04) than the older (≥65 years) cases (OR: 1.68, 95% CI: 1.02-2.76; P value for interaction: 0.006). CONCLUSIONS Despite a moderately strong association between pancreatitis (diagnosed before >2 years) and pancreatic cancer, the population attributable fraction was estimated at 1.34% (95% CI: 0.612-2.07%), suggesting that a relatively small proportion of pancreatic cancer might be avoided if pancreatitis could be prevented.
Collapse
Affiliation(s)
- E J Duell
- Unit of Nutrition, Environment and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain.
| | - E Lucenteforte
- Department of Epidemiology, Institute for Pharmacological Research 'Mario Negri', Milan; Department of Preclinical and Clinical Pharmacology 'Mario Aiazzi Mancini', University of Florence, Florence, Italy
| | - S H Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York
| | - P M Bracci
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco
| | - D Li
- Department of Gastrointestinal Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston
| | - H A Risch
- Department of Epidemiology and Public Health, School of Public Health, School of Medicine, Yale University, New Haven
| | - D T Silverman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, USA
| | - B T Ji
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, USA
| | - S Gallinger
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - E A Holly
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco
| | - E H Fontham
- School of Public Health, Louisiana State University, New Orleans, USA
| | - P Maisonneuve
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| | - H B Bueno-de-Mesquita
- National Institute for Public Health and the Environment, Bilthoven; Department of Gastroenterology and Hepatology, University Medical Centre, Utrecht, The Netherlands
| | - P Ghadirian
- Epidemiology Research Unit, Research Centre of the University of Montreal Hospital Centre (CRCHUM), Montreal, Canada
| | - R C Kurtz
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York
| | - E Ludwig
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York
| | - H Yu
- Department of Epidemiology and Public Health, School of Public Health, School of Medicine, Yale University, New Haven; Cancer Epidemiology Program, Cancer Research Center of Hawaii, University of Hawaii, Honolulu
| | - A B Lowenfels
- Department of Surgery, New York Medical College, Valhalla
| | - D Seminara
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda
| | - G M Petersen
- Departments of Health Science Research, Gastroenterology, and Medical Genetics, Mayo Clinic College of Medicine, Rochester, USA
| | - C La Vecchia
- Department of Epidemiology, Institute for Pharmacological Research 'Mario Negri', Milan; Department of Occupational Health, University of Milan, Milan, Italy
| | - P Boffetta
- The Tisch Cancer Institute and Institute for Translational Epidemiology, Mt Sinai School of Medicine, New York, USA
| |
Collapse
|
24
|
Li D, Duell EJ, Yu K, Risch HA, Olson SH, Kooperberg C, Wolpin BM, Jiao L, Dong X, Wheeler B, Arslan AA, Bueno-de-Mesquita HB, Fuchs CS, Gallinger S, Gross M, Hartge P, Hoover RN, Holly EA, Jacobs EJ, Klein AP, LaCroix A, Mandelson MT, Petersen G, Zheng W, Agalliu I, Albanes D, Boutron-Ruault MC, Bracci PM, Buring JE, Canzian F, Chang K, Chanock SJ, Cotterchio M, Gaziano J, Giovannucci EL, Goggins M, Hallmans G, Hankinson SE, Hoffman Bolton JA, Hunter DJ, Hutchinson A, Jacobs KB, Jenab M, Khaw KT, Kraft P, Krogh V, Kurtz RC, McWilliams RR, Mendelsohn JB, Patel AV, Rabe KG, Riboli E, Shu XO, Tjønneland A, Tobias GS, Trichopoulos D, Virtamo J, Visvanathan K, Watters J, Yu H, Zeleniuch-Jacquotte A, Amundadottir L, Stolzenberg-Solomon RZ. Pathway analysis of genome-wide association study data highlights pancreatic development genes as susceptibility factors for pancreatic cancer. Carcinogenesis 2012; 33:1384-90. [PMID: 22523087 PMCID: PMC3405651 DOI: 10.1093/carcin/bgs151] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Revised: 04/02/2012] [Accepted: 03/09/2012] [Indexed: 12/20/2022] Open
Abstract
Four loci have been associated with pancreatic cancer through genome-wide association studies (GWAS). Pathway-based analysis of GWAS data is a complementary approach to identify groups of genes or biological pathways enriched with disease-associated single-nucleotide polymorphisms (SNPs) whose individual effect sizes may be too small to be detected by standard single-locus methods. We used the adaptive rank truncated product method in a pathway-based analysis of GWAS data from 3851 pancreatic cancer cases and 3934 control participants pooled from 12 cohort studies and 8 case-control studies (PanScan). We compiled 23 biological pathways hypothesized to be relevant to pancreatic cancer and observed a nominal association between pancreatic cancer and five pathways (P < 0.05), i.e. pancreatic development, Helicobacter pylori lacto/neolacto, hedgehog, Th1/Th2 immune response and apoptosis (P = 2.0 × 10(-6), 1.6 × 10(-5), 0.0019, 0.019 and 0.023, respectively). After excluding previously identified genes from the original GWAS in three pathways (NR5A2, ABO and SHH), the pancreatic development pathway remained significant (P = 8.3 × 10(-5)), whereas the others did not. The most significant genes (P < 0.01) in the five pathways were NR5A2, HNF1A, HNF4G and PDX1 for pancreatic development; ABO for H.pylori lacto/neolacto; SHH for hedgehog; TGFBR2 and CCL18 for Th1/Th2 immune response and MAPK8 and BCL2L11 for apoptosis. Our results provide a link between inherited variation in genes important for pancreatic development and cancer and show that pathway-based approaches to analysis of GWAS data can yield important insights into the collective role of genetic risk variants in cancer.
Collapse
Affiliation(s)
| | - Eric J. Duell
- Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | | | - Sara H. Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Brian M. Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Li Jiao
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | - Bill Wheeler
- Information Management Services, Silver Spring, MD, USA
| | - Alan A. Arslan
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY, USA
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
- New York University Cancer Institute, New York, NY, USA
| | - H. Bas Bueno-de-Mesquita
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Charles S. Fuchs
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Steven Gallinger
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Canada
| | - Myron Gross
- Department of Laboratory Medicine/Pathology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Patricia Hartge
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Robert N. Hoover
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Elizabeth A. Holly
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Eric J. Jacobs
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA
| | - Alison P. Klein
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, The Bloomberg School of Public Health, The Sol Goldman Pancreatic Research Center, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Andrea LaCroix
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Margaret T. Mandelson
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Group Health Center for Health Studies, Seattle, WA, USA
| | - Gloria Petersen
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, and Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | - Ilir Agalliu
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | | | - Paige M. Bracci
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Julie E. Buring
- Department of Ambulatory Care and Prevention, Harvard Medical School, Boston, MA, USA
- Divisions of Preventive Medicine and Aging, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Federico Canzian
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kenneth Chang
- Comprehensive Digestive Disease Center, University of California, Irvine Medical Center, Orange, CA, USA
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
- Core Genotyping Facility, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Michelle Cotterchio
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Prevention and Cancer Control, Cancer Care Ontario, Toronto, Ontario, Canada
| | - J.Michael Gaziano
- Physicians’ Health Study, Divisions of Aging, Cardiovascular Medicine, and Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, and Massachusetts Veterans Epidemiology Research and Information Center, Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | - Edward L. Giovannucci
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard School of Public Health, Boston, MA, USA
| | - Michael Goggins
- Departments of Oncology, Pathology and Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Göran Hallmans
- Department of Public Health and Clinical Medicine, Nutritional Research, Umeå University, Umeå, Sweden
| | - Susan E. Hankinson
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Judith A. Hoffman Bolton
- Department of Epidemiology, The Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - David J. Hunter
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Amy Hutchinson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
- Core Genotyping Facility, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
| | - Kevin B. Jacobs
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
- Core Genotyping Facility, Advanced Technology Program, SAIC-Frederick Inc., NCI-Frederick, Frederick, MD, USA
- Bioinformed Consulting Services, Gaithersburg, MD, USA
| | - Mazda Jenab
- International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, Clinical Gerontology, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Peter Kraft
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard School of Public Health, Boston, MA, USA
| | - Vittorio Krogh
- Nutritional Epidemiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Robert C. Kurtz
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | - Julie B. Mendelsohn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Alpa V. Patel
- Epidemiology Research Program, American Cancer Society, Atlanta, GA, USA
| | - Kari G. Rabe
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Elio Riboli
- Division of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, and Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
| | - Anne Tjønneland
- Institute of Cancer Epidemiology, Danish Cancer Society, Copenhagen, Denmark
| | - Geoffrey S. Tobias
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Dimitrios Trichopoulos
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Bureau of Epidemiologic Research, Academy of Athens, Athens, Greece
| | - Jarmo Virtamo
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Kala Visvanathan
- Departments of Oncology, Pathology and Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joanne Watters
- Division of Cancer Prevention and Population Control, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Herbert Yu
- Yale University School of Public Health, New Haven, CT, USA
| | - Anne Zeleniuch-Jacquotte
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
- New York University Cancer Institute, New York, NY, USA
| | - Laufey Amundadottir
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Rachael Z. Stolzenberg-Solomon
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| |
Collapse
|
25
|
Olson SH, Kurtz RC. Epidemiology of pancreatic cancer and the role of family history. J Surg Oncol 2012; 107:1-7. [PMID: 22589078 DOI: 10.1002/jso.23149] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 04/17/2012] [Indexed: 12/20/2022]
Abstract
Pancreatic cancer is a lethal disease for which only a small number of risk factors have been identified. In addition to older age, male gender, and black race, risk factors include smoking, obesity, long-standing diabetes and pancreatitis, and heavy alcohol use; allergies such as hay fever are related to lowered risk. Several genetic syndromes increase risk of pancreatic cancer. Work on more common genetic variants promises to reveal more potentially important genetic associations.
Collapse
Affiliation(s)
- Sara H Olson
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
| | | |
Collapse
|