1
|
Zhang T, Moore SC, Fu S, Wang X, Albanes D, Weinstein SJ, Yu K, Stolzenberg-Solomon RZ. Association between prediagnostic serum metabolites and pancreatic ductal adenocarcinoma risk in two prospective cohorts. Int J Cancer 2025. [PMID: 40401725 DOI: 10.1002/ijc.35479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/17/2025] [Accepted: 05/08/2025] [Indexed: 05/23/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly fatal, with incidence rising worldwide. Metabolomics may provide insight into etiology and mechanisms contributing to pancreatic carcinogenesis. We examined associations between 1483 prediagnostic (up to 24 years) serum metabolites and PDAC in nested case-control studies within a cohort of male Finnish smokers and another of American men and women (n = 732 matched pairs). We used conditional logistic regression to calculate odds ratios (OR) and 95% confidence intervals per standard deviation increase in log-metabolite level within each cohort and combined using fixed-effect meta-analyses. We performed elastic net regression (EN) to select metabolites and calculated area under the curve (AUC) for established PDAC risk factors (smoking, diabetes, and overweight/obesity), selected metabolites, and their combination. Sixty-six metabolites were associated with PDAC at false discovery rate <0.05, with 26 below Bonferroni threshold (p < 3.4 × 10-5) and 38 not reported previously. Notable findings include fibrinopeptide B (1-9); 13 modified, di- or poly-peptides; 11 tobacco-chemical related xenobiotics; glycolysis-gluconeogenesis-tricarboxylic acid (TCA) cycle metabolites (aspartate, glutamate, lactate, α-ketoglutarate, and pyruvate); and four secondary and two primary bile acids that were positively (OR = 1.18-1.58) and five fibrinogen cleavage peptides that were inversely (OR = 0.70-0.84) associated with PDAC. AUCs for combined metabolites-risk factors outperformed known risk factors (p ≤ .01) but not metabolites (p ≥ .31) alone. Systemic metabolism is prospectively associated with PDAC. New metabolite associations include those related to immune response, tobacco, microbiome, glycolysis-gluconeogenesis and TCA cycle, and adiposity or diabetes. The EN selected metabolites were more sensitive indicators of prediagnostic metabolic processes and exposures associated with PDAC than established risk factors.
Collapse
Affiliation(s)
- Ting Zhang
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| | - Steven C Moore
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| | - Sheng Fu
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| | - Xiaoyu Wang
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc, Rockville, Maryland, USA
| | - Demetrius Albanes
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| | - Stephanie J Weinstein
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| | - Kai Yu
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| | - Rachael Z Stolzenberg-Solomon
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
2
|
Vermeulen R, Bodinier B, Dagnino S, Wada R, Wang X, Silverman D, Albanes D, Freedman N, Rahman M, Bell D, Chadeau-Hyam M, Rothman N. A prospective study of smoking-related white blood cell DNA methylation markers and risk of bladder cancer. Eur J Epidemiol 2024; 39:393-407. [PMID: 38554236 PMCID: PMC11101379 DOI: 10.1007/s10654-024-01110-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 02/20/2024] [Indexed: 04/01/2024]
Abstract
Bladder cancer, a common neoplasm, is primarily caused by tobacco smoking. Epigenetic alterations including DNA methylation have the potential to be used as prospective markers of increased risk, particularly in at-risk populations such as smokers. We aimed to investigate the potential of smoking-related white blood cell (WBC) methylation markers to contribute to an increase in bladder cancer risk prediction over classical questionnaire-based smoking metrics (i.e., duration, intensity, packyears) in a nested case-control study within the prospective prostate, lung, colorectal, and ovarian (PLCO) Cancer Screening Trial and the alpha-tocopherol, beta-carotene cancer (ATBC) Prevention Study (789 cases; 849 controls). We identified 200 differentially methylated sites associated with smoking status and 28 significantly associated (after correction for multiple testing) with bladder cancer risk among 2670 previously reported smoking-related cytosine-phosphate-guanines sites (CpGs). Similar patterns were observed across cohorts. Receiver operating characteristic (ROC) analyses indicated that cg05575921 (AHHR), the strongest smoking-related association we identified for bladder cancer risk, alone yielded similar predictive performance (AUC: 0.60) than classical smoking metrics (AUC: 0.59-0.62). Best prediction was achieved by including the first principal component (PC1) from the 200 smoking-related CpGs alongside smoking metrics (AUC: 0.63-0.65). Further, PC1 remained significantly associated with elevated bladder cancer risk after adjusting for smoking metrics. These findings suggest DNA methylation profiles reflect aspects of tobacco smoke exposure in addition to those captured by smoking duration, intensity and packyears, and/or individual susceptibility relevant to bladder cancer etiology, warranting further investigation.
Collapse
Affiliation(s)
- Roel Vermeulen
- Institute for Risk Assessment Sciences, Division of Environmental Epidemiology, Utrecht University, PO Box 80178, 3508 TD, Utrecht, The Netherlands.
| | - Barbara Bodinier
- Faculty of Medicine, School of Public Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC Centre for Environment and Health, Imperial College London, London, UK
| | - Sonia Dagnino
- Faculty of Medicine, School of Public Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC Centre for Environment and Health, Imperial College London, London, UK
- Commissariat À L'Energie Atomique Et Aux Énergies Alternatives (CEA), Institut Des Sciences du Vivant Fréderic Joliot, Université Côte d'Azur, Nice, France
| | - Rin Wada
- Faculty of Medicine, School of Public Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC Centre for Environment and Health, Imperial College London, London, UK
| | - Xuting Wang
- Immunity Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, RTP, Durham, NC, USA
| | - Debra Silverman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Neal Freedman
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Mohammad Rahman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Douglas Bell
- Immunity Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, RTP, Durham, NC, USA
| | - Marc Chadeau-Hyam
- Faculty of Medicine, School of Public Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Commissariat À L'Energie Atomique Et Aux Énergies Alternatives (CEA), Institut Des Sciences du Vivant Fréderic Joliot, Université Côte d'Azur, Nice, France
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
3
|
Reay WR, Kiltschewskij DJ, Di Biase MA, Gerring ZF, Kundu K, Surendran P, Greco LA, Clarke ED, Collins CE, Mondul AM, Albanes D, Cairns MJ. Genetic influences on circulating retinol and its relationship to human health. Nat Commun 2024; 15:1490. [PMID: 38374065 PMCID: PMC10876955 DOI: 10.1038/s41467-024-45779-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/04/2024] [Indexed: 02/21/2024] Open
Abstract
Retinol is a fat-soluble vitamin that plays an essential role in many biological processes throughout the human lifespan. Here, we perform the largest genome-wide association study (GWAS) of retinol to date in up to 22,274 participants. We identify eight common variant loci associated with retinol, as well as a rare-variant signal. An integrative gene prioritisation pipeline supports novel retinol-associated genes outside of the main retinol transport complex (RBP4:TTR) related to lipid biology, energy homoeostasis, and endocrine signalling. Genetic proxies of circulating retinol were then used to estimate causal relationships with almost 20,000 clinical phenotypes via a phenome-wide Mendelian randomisation study (MR-pheWAS). The MR-pheWAS suggests that retinol may exert causal effects on inflammation, adiposity, ocular measures, the microbiome, and MRI-derived brain phenotypes, amongst several others. Conversely, circulating retinol may be causally influenced by factors including lipids and serum creatinine. Finally, we demonstrate how a retinol polygenic score could identify individuals more likely to fall outside of the normative range of circulating retinol for a given age. In summary, this study provides a comprehensive evaluation of the genetics of circulating retinol, as well as revealing traits which should be prioritised for further investigation with respect to retinol related therapies or nutritional intervention.
Collapse
Affiliation(s)
- William R Reay
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia.
| | - Dylan J Kiltschewskij
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Maria A Di Biase
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zachary F Gerring
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kousik Kundu
- Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
| | - Laura A Greco
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Erin D Clarke
- School of Health Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Food and Nutrition Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Clare E Collins
- School of Health Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Food and Nutrition Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Alison M Mondul
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, Bethesda, MD, USA
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.
| |
Collapse
|
4
|
Chang VC, Rhee J, Berndt SI, Moore SC, Freedman ND, Jones RR, Silverman DT, Gierach GL, Hofmann JN, Purdue MP. Serum perfluorooctane sulfonate and perfluorooctanoate and risk of postmenopausal breast cancer according to hormone receptor status: An analysis in the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial. Int J Cancer 2023; 153:775-782. [PMID: 36843273 PMCID: PMC10405832 DOI: 10.1002/ijc.34487] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/27/2023] [Accepted: 02/16/2023] [Indexed: 02/28/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are highly persistent endocrine-disrupting chemicals that may contribute to breast cancer development; however, epidemiologic evidence is limited. We investigated associations between prediagnostic serum levels of perfluorooctane sulfonate (PFOS) and perfluorooctanoate (PFOA) and postmenopausal breast cancer risk, overall and by hormone receptor status, in a nested case-control study of 621 cases and 621 matched controls in the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial. PFOS and PFOA levels were determined based on serum metabolomic profiling performed using ultraperformance liquid chromatography-tandem mass spectrometry. We used multivariable conditional logistic regression to estimate odds ratios (ORs) and 95% confidence intervals (CIs) for the association between each PFAS and breast cancer risk, overall, by estrogen receptor (ER) or progesterone receptor (PR) status, and by joint ER/PR status. We found little evidence of association between PFOS or PFOA and breast cancer risk overall. However, in subtype-specific analyses, we observed statistically significant increased risks of ER+, PR+, and ER+/PR+ tumors for the third vs lowest quartile of serum PFOS (ORs [95% CIs] = 1.59 [1.01-2.50], 2.34 [1.29-4.23], and 2.19 [1.21-3.98], respectively) and elevated but nonstatistically significant ORs for the fourth quartile. Conversely, for PFOA, modest positive associations with ER-, PR-, ER+/PR-, and ER-/PR- tumors were generally seen in the upper quartiles. Our findings contribute evidence supporting positive associations between serum PFOS and hormone receptor-positive tumors, and possibly between PFOA and receptor-negative tumors. Future prospective studies incorporating tumor hormone receptor status are needed to better understand the role of PFAS in breast cancer etiology.
Collapse
Affiliation(s)
- Vicky C. Chang
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Jongeun Rhee
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Sonja I. Berndt
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Steven C. Moore
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Neal D. Freedman
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Rena R. Jones
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Debra T. Silverman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Gretchen L. Gierach
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Jonathan N. Hofmann
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark P. Purdue
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Zhang T, Naudin S, Hong HG, Albanes D, Männistö S, Weinstein SJ, Moore SC, Stolzenberg-Solomon RZ. Dietary Quality and Circulating Lipidomic Profiles in 2 Cohorts of Middle-Aged and Older Male Finnish Smokers and American Populations. J Nutr 2023; 153:2389-2400. [PMID: 37328109 PMCID: PMC10493471 DOI: 10.1016/j.tjnut.2023.06.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Higher dietary quality is associated with lower disease risks and has not been examined extensively with lipidomic profiles. OBJECTIVES Our goal was to examine associations of the Healthy Eating Index (HEI)-2015, Alternate HEI-2010 (AHEI-2010), and alternate Mediterranean Diet Index (aMED) diet quality indices with serum lipidomic profiles. METHODS We conducted a cross-sectional analysis of HEI-2015, AHEI-2010, and aMED with lipidomic profiles from 2 nested case-control studies within the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial (n = 627) and the Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study (n = 711). We used multivariable linear regression to determine associations of the indices, derived from baseline food-frequency questionnaires (Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial: 1993-2001, Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study: 1985-1988) with serum concentrations of 904 lipid species and 252 fatty acids (FAs) across 15 lipid classes and 28 total FAs, within each cohort and meta-analyzed results using fixed-effect models for lipids significant at Bonferroni-corrected threshold in common in both cohorts. RESULTS Adherence to HEI-2015, AHEI-2010, or aMED was associated positively with 31, 41, and 54 lipid species and 8, 6, and 10 class-specific FAs and inversely with 2, 8, and 34 lipid species and 1, 3, and 5 class-specific FAs, respectively. Twenty-five lipid species and 5 class-specific FAs were common to all indices, predominantly triacylglycerols, FA22:6 [docosahexaenoic acid (DHA)]-containing species, and DHA. All indices were positively associated with total FA22:6. AHEI-2010 and aMED were inversely associated with total FA18:1 (oleic acid) and total FA17:0 (margaric acid), respectively. The identified lipids were most associated with components of seafood and plant proteins and unsaturated:saturated fat ratio in HEI-2015; eicosapentaenoic acid plus DHA in AHEI-2010; and fish and monounsaturated:saturated fat ratio in aMED. CONCLUSIONS Adherence to HEI-2015, AHEI-2010, and aMED is associated with serum lipidomic profiles, mostly triacylglycerols or FA22:6-containing species, which are related to seafood and plant proteins, eicosapentaenoic acid-DHA, fish, or fat ratio index components.
Collapse
Affiliation(s)
- Ting Zhang
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Rockville, MD, United States
| | - Sabine Naudin
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Rockville, MD, United States; Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Hyokyoung G Hong
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Rockville, MD, United States
| | - Demetrius Albanes
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Rockville, MD, United States
| | - Satu Männistö
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Stephanie J Weinstein
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Rockville, MD, United States
| | - Steven C Moore
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Rockville, MD, United States
| | - Rachael Z Stolzenberg-Solomon
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Rockville, MD, United States.
| |
Collapse
|
6
|
Machiela MJ, Huang WY, Wong W, Berndt SI, Sampson J, De Almeida J, Abubakar M, Hislop J, Chen KL, Dagnall C, Diaz-Mayoral N, Ferrell M, Furr M, Gonzalez A, Hicks B, Hubbard AK, Hutchinson A, Jiang K, Jones K, Liu J, Loftfield E, Loukissas J, Mabie J, Merkle S, Miller E, Minasian LM, Nordgren E, Park B, Pinsky P, Riley T, Sandoval L, Saxena N, Vogt A, Wang J, Williams C, Wright P, Yeager M, Zhu B, Zhu C, Chanock SJ, Garcia-Closas M, Freedman ND. GWAS Explorer: an open-source tool to explore, visualize, and access GWAS summary statistics in the PLCO Atlas. Sci Data 2023; 10:25. [PMID: 36635305 PMCID: PMC9837135 DOI: 10.1038/s41597-022-01921-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 12/21/2022] [Indexed: 01/13/2023] Open
Abstract
The Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial is a prospective cohort study of nearly 155,000 U.S. volunteers aged 55-74 at enrollment in 1993-2001. We developed the PLCO Atlas Project, a large resource for multi-trait genome-wide association studies (GWAS), by genotyping participants with available DNA and genomic consent. Genotyping on high-density arrays and imputation was performed, and GWAS were conducted using a custom semi-automated pipeline. Association summary statistics were generated from a total of 110,562 participants of European, African and Asian ancestry. Application programming interfaces (APIs) and open-source software development kits (SKDs) enable exploring, visualizing and open data access through the PLCO Atlas GWAS Explorer website, promoting Findable, Accessible, Interoperable, and Re-usable (FAIR) principles. Currently the GWAS Explorer hosts association data for 90 traits and >78,000,000 genomic markers, focusing on cancer and cancer-related phenotypes. New traits will be posted as association data becomes available. The PLCO Atlas is a FAIR resource of high-quality genetic and phenotypic data with many potential reuse opportunities for cancer research and genetic epidemiology.
Collapse
Affiliation(s)
- Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA.
| | - Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA
| | - Wendy Wong
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA
| | - Joshua Sampson
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA
| | - Jonas De Almeida
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA
| | - Mustapha Abubakar
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA
| | - Jada Hislop
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA
| | - Kai-Ling Chen
- Essential Software Inc., Center for Biomedical Informatics and Information Technology, NCI, Rockville, USA
| | - Casey Dagnall
- Cancer Genomics Research Laboratory, DCEG, NCI, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research, Inc., Rockville, USA
| | - Norma Diaz-Mayoral
- BioProcessing and Trial Logistics Laboratory, FNLCR, Leidos Biomedical Research, Inc. Division of Cancer Prevention, NCI, NIH, Rockville, USA
| | - Mary Ferrell
- NCI at Frederick Central Repository, American Type Culture Collection, Rockville, USA
| | - Michael Furr
- Information Management Services, Inc., Danbury, USA
| | - Alex Gonzalez
- NCI at Frederick Central Repository, American Type Culture Collection, Rockville, USA
| | - Belynda Hicks
- Cancer Genomics Research Laboratory, DCEG, NCI, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research, Inc., Rockville, USA
| | - Aubrey K Hubbard
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA
| | - Amy Hutchinson
- Cancer Genomics Research Laboratory, DCEG, NCI, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research, Inc., Rockville, USA
| | - Kevin Jiang
- Essential Software Inc., Center for Biomedical Informatics and Information Technology, NCI, Rockville, USA
| | - Kristine Jones
- Cancer Genomics Research Laboratory, DCEG, NCI, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research, Inc., Rockville, USA
| | - Jia Liu
- Cancer Genomics Research Laboratory, DCEG, NCI, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research, Inc., Rockville, USA
| | - Erikka Loftfield
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA
| | - Jennifer Loukissas
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA
| | - Jerome Mabie
- Information Management Services, Inc., Danbury, USA
| | | | - Eric Miller
- Division of Cancer Prevention, NCI, NIH, Rockville, USA
| | | | - Ellen Nordgren
- NCI at Frederick Central Repository, American Type Culture Collection, Rockville, USA
| | - Brian Park
- Essential Software Inc., Center for Biomedical Informatics and Information Technology, NCI, Rockville, USA
| | - Paul Pinsky
- Division of Cancer Prevention, NCI, NIH, Rockville, USA
| | - Thomas Riley
- Information Management Services, Inc., Danbury, USA
| | - Lorena Sandoval
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA
| | - Neeraj Saxena
- Division of Cancer Prevention, NCI, NIH, Rockville, USA
| | - Aurelie Vogt
- Cancer Genomics Research Laboratory, DCEG, NCI, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research, Inc., Rockville, USA
| | - Jiahui Wang
- Cancer Genomics Research Laboratory, DCEG, NCI, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research, Inc., Rockville, USA
| | | | | | - Meredith Yeager
- Cancer Genomics Research Laboratory, DCEG, NCI, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research, Inc., Rockville, USA
| | - Bin Zhu
- Cancer Genomics Research Laboratory, DCEG, NCI, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research, Inc., Rockville, USA
| | - Claire Zhu
- Division of Cancer Prevention, NCI, NIH, Rockville, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA
| | - Montserrat Garcia-Closas
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Rockville, USA
| |
Collapse
|
7
|
King MLA, Wentzensen DN, Purdue DMP, Katki DHA, Pinto DLA, Trabert DB. Inflammatory markers in women with reported benign gynecologic pathology: An analysis of the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial. Ann Epidemiol 2021; 68:1-8. [PMID: 34906633 DOI: 10.1016/j.annepidem.2021.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 11/14/2021] [Accepted: 12/01/2021] [Indexed: 11/01/2022]
Abstract
BACKGROUND Associations between benign gynecologic pathologies and circulating inflammatory markers are unknown. Our goal was to evaluate self-reported history of benign gynecologic pathology and subsequent alterations in systemic inflammation. METHODS Using nested case-control studies from the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial, study-specific associations between self-reported history of benign ovarian cysts, uterine fibroids, and endometriosis with inflammatory marker concentrations were evaluated using logistic regression and combined using meta-analysis. Inflammatory markers associated with individual benign pathologies were mutually adjusted for one another to evaluate independent associations. RESULTS Compared to women without a self-reported history of the pathology evaluated, benign ovarian cysts were associated with increased PAI-1 (OR [95% CI] 6.24 [2.53-15.39], P<0.001) and TGF-β1 (3.79 [1.62-8.86], P=0.002) and decreased BCA-1 (0.38 [0.19-0.73], P=0.004). Uterine fibroids were associated with decreased CXCL11 (0.37 [0.22-0.63], P<0.001) and VEGFR3 (0.40 [0.24-0.65], P<0.001). Endometriosis was associated with increased SIL-4R (4.75 [1.84-12.26], P=0.001). CONCLUSIONS Self-reported history of benign gynecologic pathologies were associated with alterations in inflammatory markers that have been previously linked to cancer risk. Understanding interactions between benign gynecologic pathologies and the systemic immune system may help inform disease risk later in life.
Collapse
Affiliation(s)
- Ms Lauren A King
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, MD; University of Virginia School of Medicine, Charlottesville, VA.
| | - Dr Nicolas Wentzensen
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, MD
| | - Dr Mark P Purdue
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, MD
| | - Dr Hormuzd A Katki
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, MD
| | - Dr Ligia A Pinto
- National Cancer Institute, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Dr Britton Trabert
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, MD
| |
Collapse
|
8
|
Stolzenberg-Solomon RZ, Derkach A, Moore S, Weinstein S, Albanes D, Sampson J. Associations between metabolites and pancreatic cancer risk in a large prospective epidemiological study. Gut 2020; 69:2008-2015. [PMID: 32060129 PMCID: PMC7980697 DOI: 10.1136/gutjnl-2019-319811] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/17/2020] [Accepted: 01/20/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To assess whether prediagnostic metabolites were associated with incident pancreatic ductal adenocarcinoma (PDAC) in a prospective cohort study. DESIGN We conducted an untargeted analysis of 554 known metabolites measured in prediagnostic serum (up to 24 years) to determine their association with incident PDAC in a nested case-control study of male smokers (372 matched case-control sets) and an independent nested case-control study that included women and non-smokers (107 matched sets). Metabolites were measured using Orbitrap Elite or Q-Exactive high-resolution/accurate mass spectrometers. Controls were matched to cases by age, sex, race, date of blood draw, and follow-up time. We used conditional logistic regression adjusted for age to calculate ORs and 95% CIs for a 1 SD increase in log-metabolite level separately in each cohort and combined the two ORs using a fixed-effects meta-analysis. RESULTS Thirty-one metabolites were significantly associated with PDAC at a false discovery rate <0.05 with 12 metabolites below the Bonferroni-corrected threshold (p<9.04×10-5). Similar associations were observed in both cohorts. The dipeptides glycylvaline, aspartylphenylalanine, pyroglutamylglycine, phenylalanylphenylalanine, phenylalanylleucine and tryptophylglutamate and amino acids aspartate and glutamate were positively while the dipeptides tyrosylglutamine and α-glutamyltyrosine, fibrinogen cleavage peptide DSGEGDFXAEGGGVR and glutathione-related amino acid cysteine-glutathione disulfide were inversely associated with PDAC after Bonferroni correction. Five top metabolites demonstrated significant time-varying associations (p<0.023) with the strongest associations observed 10-15 years after participants' blood collection and attenuated thereafter. CONCLUSION Our results suggest that prediagnostic metabolites related to subclinical disease, γ-glutamyl cycle metabolism and adiposity/insulin resistance are associated with PDAC.
Collapse
Affiliation(s)
- Rachael Z. Stolzenberg-Solomon
- Metabolic Epidemiology Branch, Division Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, United States
| | - Andriy Derkach
- Biostatistics Branch, Division Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, United States
| | - Steven Moore
- Metabolic Epidemiology Branch, Division Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, United States
| | - Stephanie Weinstein
- Metabolic Epidemiology Branch, Division Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, United States
| | - Demetrius Albanes
- Metabolic Epidemiology Branch, Division Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, United States
| | - Joshua Sampson
- Biostatistics Branch, Division Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, United States
| |
Collapse
|
9
|
Huang WY, Berndt SI, Shiels MS, Katki HA, Chaturvedi AK, Wentzensen N, Trabert B, Kemp TJ, Pinto LA, Hildesheim A, Rothman N, Purdue MP. Circulating inflammation markers and colorectal adenoma risk. Carcinogenesis 2020; 40:765-770. [PMID: 30753331 DOI: 10.1093/carcin/bgz027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/28/2019] [Accepted: 02/06/2019] [Indexed: 01/10/2023] Open
Abstract
Inflammation is a driver of colorectal neoplasia; however, what particular inflammatory processes play a role in early carcinogenesis are unclear. We compared serum levels of 78 inflammation markers between 171 pathologically confirmed colorectal adenoma cases (including 48 incident cases) and 344 controls within the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial. We used weighted multivariable logistic regression to compute odds ratio (OR) and 95% confidence interval (CI). We found 14 markers associated with risk of adenoma overall; three of these were also associated with incident adenoma: CC-chemokine cysteine motif chemokine ligand 20 (CCL20) [overall adenoma fourth versus first quartile: OR 4.8, 95% CI 2.0-12, Ptrend 0.0007; incident adenoma third versus first tertile: OR 4.6, 95% CI 1.0-22, Ptrend 0.03], growth-related gene oncogene products (GRO) [OR 3.8, 95% CI 1.6-9.3, Ptrend 0.006 and OR 3.6, 95% CI 1.1-12, Ptrend 0.04, respectively] and insulin [OR 2.9, 95% CI 0.8-10, Ptrend 0.05 and OR 7.8, 95% CI 1.3-46, Ptrend 0.03, respectively]. All statistical tests were two-sided. These results provide important new evidence implicating CCL20- and GRO-related pathways in early colorectal carcinogenesis and further support a role for insulin.
Collapse
Affiliation(s)
- Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Meredith S Shiels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Hormuzd A Katki
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Anil K Chaturvedi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Troy J Kemp
- HPV Immunology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Ligia A Pinto
- HPV Immunology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Mark P Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| |
Collapse
|
10
|
Omofuma OO, Turner DP, Peterson LL, Merchant AT, Zhang J, Steck SE. Dietary Advanced Glycation End-products (AGE) and Risk of Breast Cancer in the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial (PLCO). Cancer Prev Res (Phila) 2020; 13:601-610. [PMID: 32169887 PMCID: PMC7335328 DOI: 10.1158/1940-6207.capr-19-0457] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 02/03/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022]
Abstract
Advanced glycation end-products (AGEs) are implicated in the pathogenesis of several chronic diseases including cancer. AGEs are produced endogenously but can also be consumed from foods. AGE formation in food is accelerated during cooking at high temperatures. Certain high fat or highly processed foods have high AGE values. The objective of the study was to assign and quantify Nϵ-carboxymethyl-lysine (CML)-AGE content in food and investigate the association between dietary AGE intake and breast cancer risk in the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial. The study included women enrolled in the intervention arm who were cancer-free at baseline and completed a baseline questionnaire and food frequency questionnaire (DQX). CML-AGE values were assigned and quantified to foods in the DQX using a published AGE database. Cox proportional hazards models were used to estimate the hazard ratios (HR) and 95% confidence intervals (CI) of breast cancer among all women, and stratified by race/ethnicity, invasiveness of disease, and hormone receptor status. After a median 11.5 years of follow-up, 1,592 women were diagnosed with breast cancer. Higher CML-AGE intake was associated with increased risk of breast cancer among all women (HRQ5VSQ1, 1.30; 95% CI, 1.04-1.62; P trend = 0.04) and in non-Hispanic white women (HRT3VST1, 1.21; 95% CI, 1.02-1.44). Increased CML-AGE intake was associated with increased risk of in situ (HRT3VST1, 1.49; 95% CI, 1.11-2.01) and hormone receptor-positive (HRT3VST1, 1.24; 95% CI, 1.01-1.53) breast cancers. In conclusion, high intake of dietary AGE may contribute to increased breast cancer.
Collapse
Affiliation(s)
- Omonefe O Omofuma
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
| | - David P Turner
- Medical University of South Carolina, Charleston, South Carolina
| | - Lindsay L Peterson
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Anwar T Merchant
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
| | - Jiajia Zhang
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
| | - Susan E Steck
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina.
| |
Collapse
|
11
|
Trabert B, Waterboer T, Idahl A, Brenner N, Brinton LA, Butt J, Coburn SB, Hartge P, Hufnagel K, Inturrisi F, Lissowska J, Mentzer A, Peplonska B, Sherman ME, Wills GS, Woodhall SC, Pawlita M, Wentzensen N. Antibodies Against Chlamydia trachomatis and Ovarian Cancer Risk in Two Independent Populations. J Natl Cancer Inst 2020; 111:129-136. [PMID: 29790947 DOI: 10.1093/jnci/djy084] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/20/2018] [Accepted: 04/03/2018] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Pelvic inflammatory disease (PID) has been associated with ovarian cancer risk. To clarify the role of Chlamydia trachomatis and other infectious agents in the development of ovarian cancer, we evaluated the association of serologic markers with incident ovarian cancer using a staged approach in two independent populations. METHODS Studies included: 1) a case-control study in Poland (244 ovarian cancers/556 control subjects) and 2) a prospective nested case-control study in the PLCO Cancer Screening Trial (160 ovarian cancers/159 control subjects). Associations of serologic marker levels with ovarian cancer risk at diagnostic as well as higher thresholds, identified in Poland and independently evaluated in PLCO, were estimated using multivariable adjusted logistic regression. RESULTS In the Polish study, antibodies (based on laboratory cut-point) against the chlamydia plasmid-encoded Pgp3 protein (serological gold standard) were associated with increased ovarian cancer risk (adjusted odds ratio [OR] = 1.63, 95% confidence interval [CI] = 1.20 to 2.22); when a positive result was redefined at higher levels, ovarian cancer risk was increased (cut-point 2: OR = 2.00, 95% CI = 1.38 to 2.89; cut-point 3 [max OR]: OR = 2.19, 95% CI = 1.29 to 3.73). In the prospective PLCO study, Pgp3 antibodies were associated with elevated risk at the laboratory cut-point (OR = 1.43, 95% CI = 0.78 to 2.63) and more stringent cut-points (cut-point 2: OR = 2.25, 95% CI = 1.07 to 4.71); cut-point 3: OR = 2.53, 95% CI = 0.63 to 10.08). In both studies, antibodies against other infectious agents measured were not associated with risk. CONCLUSIONS In two independent populations, antibodies against prior/current C. trachomatis (Pgp3) were associated with a doubling in ovarian cancer risk, whereas markers of other infectious agents were unrelated. These findings lend support for an association between PID and ovarian cancer.
Collapse
Affiliation(s)
- Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Tim Waterboer
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Annika Idahl
- Department of Clinical Science, Obstetrics and Gynecology, Umeå University, Umeå, Sweden
| | - Nicole Brenner
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Louise A Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Julia Butt
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sally B Coburn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Patricia Hartge
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Katrin Hufnagel
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Federica Inturrisi
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jolanta Lissowska
- Department of Epidemiology and Cancer Prevention, Cancer Center and M. Sklodowska-Curie Institute of Oncology, Warsaw, Poland
| | | | - Beata Peplonska
- Department of Environmental Epidemiology, Nofer Institute of Occupational Medicine, Lodz, Poland
| | - Mark E Sherman
- Department of Pulmonary Medicine, Mayo Clinic, Jacksonville, FL
| | - Gillian S Wills
- Jefferiss Research Trust Laboratories, Imperial College London, St Mary's Campus, London, UK
| | - Sarah C Woodhall
- National Infection Service, Public Health England, London, UK.,Research Department of Infection and Population Health, UCL, London, UK.,Gloucestershire Hospitals NHS Foundation Trust, Cheltenham, UK
| | - Michael Pawlita
- Molecular Diagnostics of Oncogenic Infections Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
12
|
Yao S, Dong SS, Ding JM, Rong Y, Zhang YJ, Chen H, Chen JB, Chen YX, Yan H, Dai Z, Guo Y. Sex-specific SNP-SNP interaction analyses within topologically associated domains reveals ANGPT1 as a novel tumor suppressor gene for lung cancer. Genes Chromosomes Cancer 2020; 59:13-22. [PMID: 31385379 DOI: 10.1002/gcc.22793] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 07/16/2019] [Accepted: 07/22/2019] [Indexed: 01/24/2023] Open
Abstract
Genetic interaction has been recognized to be an important cause of the missing heritability. The topologically associating domain (TAD) is a self-interacting genomic region, and the DNA sequences within a TAD physically interact with each other more frequently. Sex differences influence cancer susceptibility at the genetic level. Here, we performed both regular and sex-specific genetic interaction analyses within TAD to identify susceptibility genes for lung cancer in 5204 lung cancer patients and 7389 controls. We found that one SNP pair, rs4262299-rs1654701, was associated with lung cancer in women after multiple testing corrections (combined P = 8.52 × 10-9 ). Single-SNP analyses did not detect significant association signals for these two SNPs. Both identified SNPs are located in the intron region of ANGPT1. We further found that 5% of nonsmall cell lung cancer patients have an alteration in ANGPT1, indicated the potential role of ANGPT1 in the neoplastic progression in lung cancer. The expression of ANGPT1 was significantly down-regulated in patients in lung squamous cell carcinoma and lung adenocarcinoma. We checked the interaction effect on the ANGPT1 expression and lung cancer and found that the minor allele "G" of rs1654701 increased ANGPT1 gene expression and decreased lung cancer risk with the increased dosage of "A" of rs4262299, which consistent with the tumor suppressor function of ANGPT1. Survival analyses found that the high expression of ANGPT1 was individually associated with a higher survival probability in lung cancer patients. In summary, our results suggest that ANGPT1 may be a novel tumor suppressor gene for lung cancer.
Collapse
Affiliation(s)
- Shi Yao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Shan-Shan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Jing-Miao Ding
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Yu Rong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Yu-Jie Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Hao Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Jia-Bin Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Yi-Xiao Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Han Yan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Zhijun Dai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, P. R. China
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| |
Collapse
|
13
|
Muskens IS, Zhou M, Mccoy L, Bracci PM, Hansen HM, Gauderman WJ, Wiencke JK, Wrensch MR, Wiemels JL. Immune factors preceding diagnosis of glioma: a Prostate Lung Colorectal Ovarian Cancer Screening Trial nested case-control study. Neurooncol Adv 2019; 1:vdz031. [PMID: 31807733 PMCID: PMC6881819 DOI: 10.1093/noajnl/vdz031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Epidemiological studies of adult glioma have identified genetic and environmental risk factors, but much remains unclear. The aim of the current study was to evaluate anthropometric, disease-related, and prediagnostic immune-related factors for relationship with glioma risk. Methods We conducted a nested case–control study among the intervention arm of the Prostate, Lung, Colorectal, and Ovarian Cancer (PLCO) Screening Trial. One hundred and twenty-four glioma cases were identified and each matched to four controls. Baseline characteristics were collected at enrollment and were evaluated for association with glioma status. Serum specimens were collected at yearly intervals and were analyzed for immune-related factors including TGF-β1, TNF-α, total IgE, and allergen-specific IgE. Immune factors were evaluated at baseline in a multivariate conditional logistic regression model, along with one additional model that incorporated the latest available measurement. Results A family history of glioma among first-degree relatives was associated with increased glioma risk (OR = 4.41, P = .002). In multivariate modeling of immune factors at baseline, increased respiratory allergen-specific IgE was inversely associated with glioma risk (OR for allergen-specific IgE > 0.35 PAU/L: 0.59, P = .03). A logistic regression model that incorporated the latest available measurements found a similar association for allergen-specific IgE (P = .005) and showed that elevated TGF-β1 was associated with increased glioma risk (P-value for trend <.0001). Conclusion The results from this prospective prediagnostic study suggest that several immune-related factors are associated with glioma risk. The association observed for TGF-β1 when sampling closer to the time of diagnosis may reflect the nascent brain tumor’s feedback on immune function.
Collapse
Affiliation(s)
- Ivo S Muskens
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Mi Zhou
- Department of Epidemiology and Biostatistics
| | - Lucie Mccoy
- Department of Neurological Surgery, School of Medicine, University of California, San Francisco, San Francisco, CA
| | | | - Helen M Hansen
- Department of Neurological Surgery, School of Medicine, University of California, San Francisco, San Francisco, CA
| | - W James Gauderman
- Division of Biostatistics, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - John K Wiencke
- Department of Neurological Surgery, School of Medicine, University of California, San Francisco, San Francisco, CA
| | - Margaret R Wrensch
- Department of Neurological Surgery, School of Medicine, University of California, San Francisco, San Francisco, CA
| | - Joseph L Wiemels
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA.,Department of Epidemiology and Biostatistics
| |
Collapse
|
14
|
Agalliu I, Chen Z, Wang T, Hayes RB, Freedman ND, Gapstur SM, Burk RD. Oral Alpha, Beta, and Gamma HPV Types and Risk of Incident Esophageal Cancer. Cancer Epidemiol Biomarkers Prev 2018; 27:1168-1175. [PMID: 30087123 PMCID: PMC6170688 DOI: 10.1158/1055-9965.epi-18-0287] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/10/2018] [Accepted: 07/31/2018] [Indexed: 12/31/2022] Open
Abstract
Background: Several studies have examined association between human papillomaviruses (HPV) and esophageal cancer, but results have been inconsistent. This is the first prospective study to investigate associations between α, β and γ HPV detection in the oral cavity and risk of esophageal cancer.Methods: We conducted a nested case-control study among 96,650 cancer-free participants in the American Cancer Society Cancer Prevention Cohort and the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. Incident esophageal cancer cases (n = 125) were identified during an average 3.9 years of follow-up. Three controls per case (n = 372) were selected and matched on age, sex, race/ethnicity, and time since mouthwash collection. α, β, and γ HPV DNA in oral samples were detected using a next-generation sequencing assay. Conditional logistic regression models were used to estimate OR and 95% confidence intervals (CIs), adjusting for smoking and alcohol consumption. Statistical significance was evaluated using permutation test.Results: Prevalence of oral α, β, and γ HPV was 18.4%, 64.8%, and 42.4% in cases and 14.3%, 55.1%, and 33.6% in controls, respectively. Oral HPV16 detection was not associated with esophageal cancer (OR = 0.54, 95% CI, 0.1-4.84) and none of the esophageal squamous cell carcinoma cases (n = 28) were HPV16 positive. Some oral HPV types were more common in cases than controls; however, none of the associations were statistically significant.Conclusions: Although HPVs in the oral cavity are very common, this study showed no evidence of association between oral HPVs and esophageal cancer.Impact: Oral HPVs may not contribute to risk of esophageal cancer. Cancer Epidemiol Biomarkers Prev; 27(10); 1168-75. ©2018 AACR.
Collapse
Affiliation(s)
- Ilir Agalliu
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York.
| | - Zigui Chen
- Department of Pediatrics (Genetics), Albert Einstein College of Medicine, Bronx, New York
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Richard B Hayes
- Department of Population Health and Environmental Medicine, New York University, New York, New York
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Robert D Burk
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York.
- Department of Pediatrics (Genetics), Albert Einstein College of Medicine, Bronx, New York
- Departments of Microbiology and Immunology; and Obstetrics, Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
15
|
Hada M, Edin ML, Hartge P, Lih FB, Wentzensen N, Zeldin DC, Trabert B. Prediagnostic Serum Levels of Fatty Acid Metabolites and Risk of Ovarian Cancer in the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. Cancer Epidemiol Biomarkers Prev 2018; 28:189-197. [PMID: 30262599 DOI: 10.1158/1055-9965.epi-18-0392] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/10/2018] [Accepted: 09/19/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Evidence suggests that inflammation increases risk for ovarian cancer. Aspirin has been shown to decrease ovarian cancer risk, though the mechanism is unknown. Studies of inflammatory markers, lipid molecules such as arachidonic acid, linoleic acid, and alpha-linoleic acid metabolites, and development of ovarian cancer are essential to understand the potential mechanisms. METHODS We conducted a nested case-control study (157 cases/156 matched controls) within the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. Unconditional logistic regression was used to estimate the association between prediagnostic serum levels of 31 arachidonic acid/linoleic acid/alpha-linoleic acid metabolites and risk of ovarian cancer. RESULTS Five of the 31 arachidonic acid/linoleic acid/alpha-linoleic acid (free fatty acids) metabolites were positively associated with ovarian cancer risk: 8-HETE [tertile 3 vs. 1: OR 2.53 (95% confidence interval [CI] 1.18-5.39), P trend 0.02], 12,13-DHOME [2.49 (1.29-4.81), 0.01], 13-HODE [2.47 (1.32-4.60), 0.005], 9-HODE [1.97 (1.06-3.68), 0.03], 9,12,13-THOME [2.25 (1.20-4.21), 0.01]. In analyses by subtype, heterogeneity was suggested for 8-HETE [serous OR (95% CI): 2.53 (1.18-5.39) vs. nonserous OR (95% CI): 1.15 (0.56-2.36), P het 0.1] and 12,13-EpOME [1.95 (0.90-4.22) vs. 0.82 (0.39-1.73), 0.05]. CONCLUSIONS Women with increased levels of five fatty acid metabolites (8-HETE, 12,13-DHOME, 13-HODE, 9-HODE, and 9,12,13-THOME) were at increased risk of developing ovarian cancer in the ensuing decade. All five metabolites are derived from either arachidonic acid (8-HETE) or linoleic acid (12,13-DHOME, 13-HODE, 9-HODE, 9,12,13-THOME) via metabolism through the LOX/cytochrome P450 pathway. IMPACT The identification of these risk-related fatty acid metabolites provides mechanistic insights into the etiology of ovarian cancer and indicates the direction for future research.
Collapse
Affiliation(s)
- Manila Hada
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Matthew L Edin
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Patricia Hartge
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Fred B Lih
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Darryl C Zeldin
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
16
|
Hu B, Wang X, He JB, Dai YJ, Zhang J, Yu Y, Sun Q, Lin-FengYan, Hu YC, Nan HY, Yang Y, Kaye AD, Cui GB, Wang W. Structural and functional brain changes in perimenopausal women who are susceptible to migraine: a study protocol of multi-modal MRI trial. BMC Med Imaging 2018; 18:26. [PMID: 30189858 PMCID: PMC6127929 DOI: 10.1186/s12880-018-0272-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 08/29/2018] [Indexed: 01/01/2023] Open
Abstract
Background As a common clinical symptom that often bothers midlife females, migraine is closely associated with perimenopause. Previous studies suggest that one of the most prominent triggers is the sudden decline of estrogen during perimenopausal period. Hormone replacement therapy (HRT) is widely used to prevent this suffering in perimenopausal women, but effective diagnostic system is lacked for quantifying the severity of the diseaase. To avoid the abuse and overuse of HRT, we propose to conduct a diagnostic trial using multimodal MRI techniques to quantify the severity of these perimenopausal migraineurs who are susceptible to the decline of estrogen. Methods Perimenopausal women suffering from migraine will be recruited from the pain clinic of our hospital. Perimenopausal women not suffering from any kind of headache will be recruited from the local community. Clinical assessment and multi-modal MR imaging examination will be conducted. A follow up will be conducted once half year within 3 years. Pain behavior, neuropsychology scores, fMRI analysis combined with suitable statistical software will be used to reveal the potential association between these above traits and the susceptibility of migraine. Discussion Multi-modal imaging features of both healthy controls and perimenopausal women who are susceptible to estrogen decline will be acquired. Imaging features will include volumetric characteristics, white matter integrity, functional characteristics, topological properties, and perfusion properties. Clinical information, such as basic information, blood estrogen level, information of migraine, and a bunch of neurological scale will also be used for statistic assessment. This clinical trial would help to build an effective screen system for quantifying the severity of illness of those susceptible women during the perimenopausal period. Trial registration This study has already been registered at Clinical Trials. gov (ID: NCT02820974). Registration date: September 28th, 2014.
Collapse
Affiliation(s)
- Bo Hu
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Xu Wang
- Student Brigade, Fourth Military Medical University (Air Force Medical University), 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Jie-Bing He
- Student Brigade, Fourth Military Medical University (Air Force Medical University), 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Yu-Jie Dai
- Department of Clinical Nutrition, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jin Zhang
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Ying Yu
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Qian Sun
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Lin-FengYan
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Yu-Chuan Hu
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Hai-Yan Nan
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Yang Yang
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Alan D Kaye
- Departments of Anesthesiology and Pharmacology, Louisiana State University School of Medicine, New Orleans, Louisiana, USA
| | - Guang-Bin Cui
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| | - Wen Wang
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
17
|
Li Z, Pu Z, Fan J, Li N, Zhu M, Zhang J, Wang Y, Geng L, Cheng Y, Ma H, Jin G, Dai J, Hu Z, Shen H. Fine mapping in TERT-CLPTM1L region identified three independent lung cancer susceptibility signals: A large-scale multi-ethnic population study. Mol Carcinog 2018; 57:1289-1299. [PMID: 29809284 DOI: 10.1002/mc.22843] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 12/31/2022]
Abstract
Genome-wide association studies (GWAS) and fine mapping studies have identified multiple lung cancer susceptibility variants in TERT-CLPTM1L region. However, it is still unclear about the relationship between these risk variants and the independent lung cancer risk signals in this region. Therefore, we evaluated the independent susceptibility signals for lung cancer and explored the potential functional variants in this region. Sequential conditional analysis was used to detect the independent susceptibility loci based on four lung cancer GWAS datasets with 12 843 lung cases and 12 639 controls. Comprehensively functional annotations were performed for each independent signal. Three independent susceptibility signals were identified in multi-ethnic population. For the first signal, rs2736100 showed the most significant association with lung cancer risk (C > A, OR = 0.82, 95%CI: 0.79-0.85, P = 1.98 × 10-25 ). Rs36019446 was the top-ranked site (A > G, OR = 0.88, 95%CI: 0.84-0.92, P = 1.74 × 10-9 ) in the second signal. For the third signal, rs326048 was the leading SNP (A > G, OR = 0.91, 95%CI: 0.87-0.95, P = 1.38 × 10-5 ). The following subgroup analysis found the same three loci among Asian population. Further, we compared the difference between various subgroup populations. Functional annotations revealed that rs2736100, rs27996 (r2 = 0.85 with rs36019446) and rs326049 (r2 = 0.73 with rs326048) could be potential functional variants in these three risk signals, respectively. In conclusion, although multiple variants have been found associated with lung cancer risk in TERT-CLPTM1L region, our findings indicated that there are three independent lung cancer susceptibility signals in this region.
Collapse
Affiliation(s)
- Zhihua Li
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhening Pu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jingyi Fan
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ni Li
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meng Zhu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jiahui Zhang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yuzhuo Wang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Liguo Geng
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yang Cheng
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hongxia Ma
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Guangfu Jin
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Juncheng Dai
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Zhibin Hu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| | - Hongbing Shen
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Guinter MA, McLain AC, Merchant AT, Sandler DP, Steck SE. An estrogen-related lifestyle score is associated with risk of postmenopausal breast cancer in the PLCO cohort. Breast Cancer Res Treat 2018; 170:613-622. [PMID: 29651647 DOI: 10.1007/s10549-018-4784-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 04/07/2018] [Indexed: 01/26/2023]
Abstract
PURPOSE Healthy or unhealthy lifestyle behaviors are often adopted together. We aimed to investigate the combined effect of estrogen-related lifestyle factors on postmenopausal breast cancer risk. METHODS Data from 27,153 women enrolled in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial were used. We created an estrogen-related lifestyle score (ERLS) by incorporating a previously developed measure of estrogenic diet, alcohol intake, body mass index (BMI), and physical activity. The scores ranged from 0 to 6 with alcohol and BMI accounting for higher weights than the other factors. To evaluate the preventive possibilities of a low estrogen-related lifestyle and to be consistent with other published lifestyle scores, higher scores were set to correspond with potentially lower estrogenic lifestyle. The association between the ERLS and incident breast cancer was examined using Cox proportional hazards models. RESULTS Participants with an ERLS of 4 or ≥ 5 had a 23% (HR 0.77; 95% CI 0.67-0.89) and 34% (HR 0.66; 95% CI 0.56-0.78) lower risk of breast cancer, respectively, compared to those with an ERLS ≤ 2 after multivariable adjustment. Estimates were similar when restricting to invasive cases or estrogen receptor-positive subtypes. No single lifestyle component appeared to drive the association. CONCLUSIONS Our findings suggest that the combined effect of a lifestyle characterized by a low estrogenic diet, low alcohol consumption, low body weight, and high levels of physical activity are associated with a reduction in postmenopausal breast cancer risk, possibly through an influence on estrogen metabolism.
Collapse
Affiliation(s)
- Mark A Guinter
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, GA, USA
| | - Alexander C McLain
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Anwar T Merchant
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Susan E Steck
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA. .,Cancer Prevention and Control Program, University of South Carolina, Columbia, SC, 29208, USA.
| |
Collapse
|
19
|
Guinter MA, McLain AC, Merchant AT, Sandler DP, Steck SE. A dietary pattern based on estrogen metabolism is associated with breast cancer risk in a prospective cohort of postmenopausal women. Int J Cancer 2018; 143:580-590. [PMID: 29574860 DOI: 10.1002/ijc.31387] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 02/14/2018] [Accepted: 02/28/2018] [Indexed: 12/27/2022]
Abstract
Increased exposure to estrogen is a risk factor for postmenopausal breast cancer, and dietary factors can influence estrogen metabolism. However, studies of diet and breast cancer have been inconclusive. We developed a dietary pattern associated with levels of unconjugated estradiol and the ratio of 2- and 16-hydroxylated estrogen metabolites in a subsample of Prostate, Lung, Colorectal and Ovarian Screening Trial (PLCO) participants (n = 653) using reduced rank regression, and examined its association with postmenopausal breast cancer prospectively in the larger PLCO cohort (n = 27,488). The estrogen-related dietary pattern (ERDP) was comprised of foods with positively-weighted intakes (non-whole/refined grains, tomatoes, cruciferous vegetables, cheese, fish/shellfish high in ω-3 fatty acids, franks/luncheon meats) and negatively-weighted intakes (nuts/seeds, other vegetables, fish/shellfish low in ω-3 fatty acids, yogurt, coffee). A 1-unit increase in the ERDP score was associated with an increase in total (HR: 1.09, 95% CI: 1.01-1.18), invasive (HR: 1.13; 95% CI: 1.04-1.24) and estrogen receptor (ER)-positive (HR: 1.13, 95% CI: 1.02-1.24) breast cancer risk after adjustment for confounders. Associations were observed for the fourth quartile of ERDP compared with the first quartile for overall breast cancer (HR: 1.14; 95% CI: 0.98-1.32), invasive cases (HR: 1.20, 95% CI: 1.02-1.42) and ER-positive cases (HR: 1.19; 95% CI: 0.99-1.41). The increased risk associated with increasing ERDP score was more apparent in strata of some effect modifiers (postmenopausal hormone therapy non-users and non-obese participants) where the relative estrogen exposure due to that factor was lowest, although the p values for interaction were not statistically significant. Results suggest a dietary pattern based on estrogen metabolism is positively associated with postmenopausal breast cancer risk, possibly through an estrogenic influence.
Collapse
Affiliation(s)
- Mark A Guinter
- Behavioral and Epidemiology Research Group, American Cancer Society, Atlanta, GA.,Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - Alexander C McLain
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - Anwar T Merchant
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Susan E Steck
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC.,Cancer Prevention and Control Program, University of South Carolina, Columbia, SC
| |
Collapse
|
20
|
Abstract
Lung cancer is the leading cause of cancer deaths in both men and women in the US. While most sporadic lung cancer cases are related to environmental factors such as smoking, genetic susceptibility may also play an important role and a number of lung cancer associated single-nucleotide polymorphisms (SNPs) have been identified although many remain to be found. The collective effects of genome-wide minor alleles of common SNPs, or the minor allele content (MAC) in an individual, have been linked with quantitative variations of complex traits and diseases. Here we studied MAC in lung cancer using previously published SNPs data sets (US and Finland samples) and found higher MAC in cases relative to matched controls. A set of 5400 SNPs with MA (MAF < 0.5) more common in cases (P < 0.08) and linkage disequilibrium (LD) r2 = 0.3 was found to have the best predictive accuracy. These results identify higher MAC in lung cancer susceptibility and provide a meaningful genetic method to identify those at risk of lung cancer.
Collapse
|
21
|
White blood cell DNA methylation and risk of breast cancer in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO). Breast Cancer Res 2017; 19:94. [PMID: 28821281 PMCID: PMC5563066 DOI: 10.1186/s13058-017-0886-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/25/2017] [Indexed: 01/24/2023] Open
Abstract
Background Several studies have suggested that global DNA methylation in circulating white blood cells (WBC) is associated with breast cancer risk. Methods To address conflicting results and concerns that the findings for WBC DNA methylation in some prior studies may reflect disease effects, we evaluated the relationship between global levels of WBC DNA methylation in white blood cells and breast cancer risk in a case-control study nested within the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial (PLCO) cohort. A total of 428 invasive breast cancer cases and 419 controls, frequency matched on age at entry (55–59, 60–64, 65–69, ≥70 years), year of entry (on/before September 30, 1997, on/after October 1, 1997) and period of DNA extraction (previously extracted, newly extracted) were included. The ratio of 5-methyl-2’ deoxycytidine [5-mdC] to 2’-deoxyguanine [dG], assuming [dG] = [5-mdC] + [2’-deoxycytidine [dC]] (%5-mdC), was determined by liquid chromatography-electrospray ionization-tandem mass spectrometry, an especially accurate method for assessing total genomic DNA methylation. Results Odds ratio (OR) estimates and 95% confidence intervals (CI) for breast cancer risk adjusted for age at entry, year of entry, and period of DNA extraction, were 1.0 (referent), 0.89 (95% CI, 0.6–1.3), 0.88 (95% CI, 0.6–1.3), and 0.84 (95% CI, 0.6–1.2) for women in the highest compared to lowest quartile levels of %5md-C (p for trend = .39). Effects did not meaningfully vary by time elapsed from WBC collection to diagnosis. Discussion These results do not support the hypothesis that global DNA hypomethylation in WBC DNA is associated with increased breast cancer risk prior to the appearance of clinical disease.
Collapse
|
22
|
Playdon MC, Ziegler RG, Sampson JN, Stolzenberg-Solomon R, Thompson HJ, Irwin ML, Mayne ST, Hoover RN, Moore SC. Nutritional metabolomics and breast cancer risk in a prospective study. Am J Clin Nutr 2017; 106:637-649. [PMID: 28659298 PMCID: PMC5525118 DOI: 10.3945/ajcn.116.150912] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 05/30/2017] [Indexed: 12/16/2022] Open
Abstract
Background: The epidemiologic evidence for associations between dietary factors and breast cancer is weak and etiologic mechanisms are often unclear. Exploring the role of dietary biomarkers with metabolomics can potentially facilitate objective dietary characterization, mitigate errors related to self-reported diet, agnostically test metabolic pathways, and identify mechanistic mediators.Objective: The aim of this study was to evaluate associations of diet-related metabolites with the risk of breast cancer in the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial.Design: We examined prediagnostic serum concentrations of diet-related metabolites in a nested case-control study in 621 postmenopausal invasive breast cancer cases and 621 matched controls in the multicenter PLCO cohort. We calculated partial Pearson correlations between 617 metabolites and 55 foods, food groups, and vitamin supplements on the basis of the 2015 Dietary Guidelines for Americans and derived from a 137-item self-administered food-frequency questionnaire. Diet-related metabolites (P-correlation < 1.47 × 10-6) were evaluated in breast cancer analyses. ORs for the 90th compared with the 10th percentile were calculated by using conditional logistic regression, with body mass index, physical inactivity, other breast cancer risk factors, and caloric intake controlled for (false discovery rate <0.2).Results: Of 113 diet-related metabolites, 3 were associated with overall breast cancer risk (621 cases): caprate (10:0), a saturated fatty acid (OR: 1.77; 95% CI = 1.28, 2.43); γ-carboxyethyl hydrochroman (γ-CEHC), a vitamin E (γ-tocopherol) derivative (OR: 1.64; 95% CI: 1.18, 2.28); and 4-androsten-3β,17β-diol-monosulfate (1), an androgen (OR: 1.61; 95% CI: 1.20, 2.16). Nineteen metabolites were significantly associated with estrogen receptor (ER)-positive (ER+) breast cancer (418 cases): 12 alcohol-associated metabolites, including 7 androgens and α-hydroxyisovalerate (OR: 2.23; 95% CI: 1.50, 3.32); 3 vitamin E (tocopherol) derivatives (e.g., γ-CEHC; OR: 1.80; 95% CI: 1.20, 2.70); butter-associated caprate (10:0) (OR: 1.81; 95% CI: 1.23, 2.67); and fried food-associated 2-hydroxyoctanoate (OR: 1.46; 95% CI: 1.03, 2.07). No metabolites were significantly associated with ER-negative breast cancer (144 cases).Conclusions: Prediagnostic serum concentrations of metabolites related to alcohol, vitamin E, and animal fats were moderately strongly associated with ER+ breast cancer risk. Our findings show how nutritional metabolomics might identify diet-related exposures that modulate cancer risk. This trial was registered at clinicaltrials.gov as NCT00339495.
Collapse
Affiliation(s)
- Mary C Playdon
- Yale School of Public Health, Yale University, New Haven, CT; .,Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | - Regina G Ziegler
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | - Joshua N Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | | | - Henry J Thompson
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO
| | - Melinda L Irwin
- Yale School of Public Health, Yale University, New Haven, CT;,Yale Cancer Center, New Haven, CT; and
| | - Susan T Mayne
- Yale School of Public Health, Yale University, New Haven, CT;,US Food and Drug Administration, College Park, MD
| | - Robert N Hoover
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | - Steven C Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| |
Collapse
|
23
|
Qin N, Wang C, Lu Q, Huang T, Zhu M, Wang L, Yu F, Huang M, Jiang Y, Dai J, Ma H, Jin G, Wu C, Lin D, Shen H, Hu Z. A cis-eQTL genetic variant of the cancer-testis gene CCDC116 is associated with risk of multiple cancers. Hum Genet 2017; 136:987-997. [PMID: 28653172 DOI: 10.1007/s00439-017-1827-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/19/2017] [Indexed: 11/29/2022]
Abstract
Recent studies have found that cancer-testis (CT) genes, which are expressed predominantly in germ and cancer cells, may be candidate cancer drivers. Because of their crucial roles, genetic variants in these genes may contribute to the development of cancer. Here, we systematically evaluated associations of common variants in CT genes and their promoters for the risk of lung cancer in our initial GWAS (2331 cases and 3077 controls), followed by in silico replication using additional 10,512 lung cancer cases and 9562 controls. We found a significant association between rs3747093 located in the CCDC116 promoter and lung cancer risk (OR = 0.91, P meta = 7.81 × 10-6). Although CCDC116 was expressed at lower levels in somatic tissues compared to the testis, the protective allele A of rs3747093 was associated with decreased CCDC116 expression in many normal tissues, including the lung (P = 8.1 × 10-13). We subsequently genotyped this variant in another four commonly diagnosed cancers (gastric, esophageal, colorectal, and breast cancers), as we found expression quantitative trait locus (eQTL) signals for rs3747093 and CCDC116 in their corresponding normal tissues. Interestingly, we observed consistent associations between rs3747093 and multiple cancers (gastric cancer: OR = 0.85, P = 2.21 × 10-4; esophageal cancer: OR = 0.91, P = 2.57 × 10-2; colorectal cancer: OR = 0.80, P = 1.85 × 10-6; and breast cancer: OR = 0.87, P = 1.55 × 10-3). Taken together, the A allele of rs3747093 showed significant protective effects on cancer risk (OR = 0.88, P pool = 6.52 × 10-13) in an Asian population. Moreover, our findings suggest that low abundance expression of CT genes in normal tissues may also contribute to tumorigenesis, providing a new mechanism of CT genes in the development of cancer.
Collapse
Affiliation(s)
- Na Qin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Cheng Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Qun Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Tongtong Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Meng Zhu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Lihua Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Fei Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Mingtao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Yue Jiang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Juncheng Dai
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Hongxia Ma
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Guangfu Jin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Chen Wu
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dongxin Lin
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hongbing Shen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China.,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China. .,Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
24
|
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: 1.1] [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
|
25
|
Barry KH, Moore LE, Sampson JN, Koutros S, Yan L, Meyer A, Reddy M, Oler AJ, Cook MB, Fraumeni Jr JF, Yeager M, Amundadottir LT, Berndt SI. Prospective study of DNA methylation at chromosome 8q24 in peripheral blood and prostate cancer risk. Br J Cancer 2017; 116:1470-1479. [PMID: 28463958 PMCID: PMC5520085 DOI: 10.1038/bjc.2017.104] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/17/2017] [Accepted: 03/23/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Chromosome 8q24 has emerged as an important genetic susceptibility region for several cancers, including prostate cancer; however, little is known about the contribution of DNA methylation in this region to risk. METHODS We prospectively evaluated DNA methylation at 8q24 in relation to prostate cancer using pre-diagnostic blood samples from 694 prostate cancer cases (including 172 aggressive cases) and 703 controls in the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial. We used logistic regression to estimate odds ratios and 95% confidence intervals. RESULTS Although none remained significant after adjustment for multiple testing (q>0.05), of the 50 CpG sites meeting quality control, we identified 8 sites that were nominally associated with prostate cancer (Ptrend<0.05), including 6 correlated (Spearman ρ: 0.20-0.52) sites in POU5F1B and 2 intergenic sites (most significant site: Chr8:128428897 in POU5F1B, Ptrend=0.01). We also identified two correlated (ρ=0.39) sites in MYC (Chr8:128753187 and Chr8:128753154) that were associated with aggressive (Ptrend=0.02 and 0.03), but not non-aggressive disease (Ptrend=0.70 and 0.20; Pheterogeneity=0.01 and 4.6 × 10-3). These findings persisted after adjustment for the top 8q24 prostate cancer variants in our study. CONCLUSIONS Although requiring replication, our findings provide some evidence that 8q24 DNA methylation levels may be associated with prostate cancer risk.
Collapse
Affiliation(s)
- Kathryn Hughes Barry
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Program in Oncology, University of Maryland, Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Lee E Moore
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Joshua N Sampson
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Stella Koutros
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Liying Yan
- EpigenDx, Inc., Hopkinton, MA 01748, USA
| | - Ann Meyer
- EpigenDx, Inc., Hopkinton, MA 01748, USA
| | | | - Andrew J Oler
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Michael B Cook
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Joseph F Fraumeni Jr
- Office of the Director, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Meredith Yeager
- Frederick National Laboratory for Cancer Research, Cancer Genomics Research Laboratory, Leidos Biomedical Research, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Sonja I Berndt
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
26
|
Dong J, Cheng Y, Zhu M, Wen Y, Wang C, Wang Y, Geng L, Shen W, Liu J, Li Z, Zhang J, Ma H, Dai J, Jin G, Hu Z, Shen H. Fine mapping of chromosome 5p15.33 identifies novel lung cancer susceptibility loci in Han Chinese. Int J Cancer 2017; 141:447-456. [PMID: 28335076 DOI: 10.1002/ijc.30702] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 03/05/2017] [Accepted: 03/07/2017] [Indexed: 12/20/2022]
Abstract
Genome-wide association studies in European and Asian populations have consistently identified chromosome 5p15.33 as a lung cancer susceptibility region. To investigate further the genetic architecture of common variants in this region, we conducted a two-stage fine-mapping analysis discovered by targeted resequencing of 200 cases and 300 controls individually, and validated in multiethnic lung cancer Genome wide association studies (GWASs) with 12,843 cases and 12,639 controls. Two independent variants were identified in approximate conditional analysis with GCTA and consistently validated in lung cancer GWASs in both Asian and European populations. These were rs10054203 in TERT (resequencing: OR = 1.69, p = 2.70 × 10-4 ; validation: OR = 1.34, p = 2.10 × 10-23 for Asian, and OR = 1.09, p = 6.00 × 10-3 for European), and rs397640 in CLPTM1L (resequencing: OR = 0.37, p = 1.19 × 10-4 ; validation: OR = 0.75, p = 5.89 × 10-8 for Asian, and OR = 0.90, p = 2.40 × 10-2 for European). Expression quantitative trait loci analysis showed the risk allele (C) of rs10054203 was significantly associated with lower mRNA expression of CTD-2245Ef15.3 (p = 0.019) and Tubulin Polymerization-Promoting Protein (TPPP, p = 0.031) in 167 lung tissues. In conclusion, in this largest and first resequencing-based fine-mapping analysis of 5p15.33 region in Han Chinese, we identified two novel variants associated with lung cancer susceptibility. Further validation studies and functional work is required to confirm the roles of the newly discovered variants.
Collapse
Affiliation(s)
- Jing Dong
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yang Cheng
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Meng Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yang Wen
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Cheng Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yuzhuo Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Liguo Geng
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Wei Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jia Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Zhihua Li
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jiahui Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Hongxia Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Juncheng Dai
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Guangfu Jin
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Zhibin Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China
| |
Collapse
|
27
|
Huang WY, Kemp TJ, Pfeiffer RM, Pinto LA, Hildesheim A, Purdue MP. Impact of freeze-thaw cycles on circulating inflammation marker measurements. Cytokine 2017; 95:113-117. [PMID: 28260648 DOI: 10.1016/j.cyto.2017.02.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/18/2017] [Accepted: 02/17/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Circulating inflammation markers are being increasingly measured in prospective cohorts to investigate cancer etiology. However, it is unclear how the measurements are affected by the freeze-thaw cycles of the specimens prior to marker analysis. METHODS We compared concentrations of 45 inflammation markers between paired serum vials of 55 participants in the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial that have undergone one (T1), two (T2), and three (T3) freeze-thaw cycles at the time of assay. We computed the difference of analyte concentrations across paired vials (T1 vs. T2, T2 vs. T3) and tested whether the difference deviated from zero using the Wilcoxon signed-rank test. We also calculated Spearman rank correlation and weighted kappa statistics for T1 vs. T2 and T2 vs. T3 comparisons to assess agreement in rank ordering of subjects. RESULTS Measurements between paired T1 and T2 samples were largely similar, with the difference not statistically deviating from zero for 36 of the 45 markers. In contrast, tests of the difference between paired T2 and T3 samples were statistically significant for 36 markers. However, the rank ordering of participants by marker concentration remained largely consistent across T2 and T3 samples, with Spearman correlation coefficients >0.8 for 42 markers and weighted kappas >0.7 for 37 markers. CONCLUSION We recommend that studies measuring inflammation markers use previously unthawed specimens to the extent possible, or match on the number of prior freeze-thaw cycles in nested case-control studies.
Collapse
Affiliation(s)
- Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Rockville, MD, United States.
| | - Troy J Kemp
- HPV Immunology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Rockville, MD, United States
| | - Ligia A Pinto
- HPV Immunology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Rockville, MD, United States
| | - Mark P Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Rockville, MD, United States
| |
Collapse
|
28
|
Trabert B, Eldridge RC, Pfeiffer RM, Shiels MS, Kemp TJ, Guillemette C, Hartge P, Sherman ME, Brinton LA, Black A, Chaturvedi AK, Hildesheim A, Berndt SI, Safaeian M, Pinto L, Wentzensen N. Prediagnostic circulating inflammation markers and endometrial cancer risk in the prostate, lung, colorectal and ovarian cancer (PLCO) screening trial. Int J Cancer 2016; 140:600-610. [PMID: 27770434 DOI: 10.1002/ijc.30478] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/07/2016] [Indexed: 12/12/2022]
Abstract
Inflammation is proposed to increase risk of developing endometrial cancer, but few prospective epidemiologic studies have investigated the relationship between circulating inflammation markers and endometrial cancer risk. In a nested case-control study within the PLCO Screening Trial we measured serum levels of 64 inflammation-related biomarkers in 284 incident endometrial cancer cases and 284 matched controls. Using multivariable logistic regression inflammation markers were evaluated individually and combined into a cross-validated inflammation score. Of 64 markers, 22 were associated with endometrial cancer risk at p < 0.05 and 17 of 22 markers remained associated after multiple testing corrections. After adjusting for BMI and estradiol, SERPINE1 [quartile(Q)4 vs. Q1 odds ratio (OR) (95% confidence interval (CI)), p trend = 2.43 (0.94-6.29), 0.03] and VEGFA [2.56 (1.52-4.30), 0.0002] were positively associated with endometrial cancer risk, while CCL3 [0.46 (0.27-0.77), 0.01], IL13 [0.55 (0.33-0.93), 0.01], IL21 [0.52 (0.31-0.87), 0.01], IL1B [0.51 (0.30-0.86), 0.01] and IL23 [0.60 (0.35-1.03), 0.02] were inversely associated with risk. We observed large differences in ORs across BMI-inflammation score categories. Endometrial cancer risk was most pronounced among obese women with the highest inflammation score tertile (T) [10.25 (3.56-29.55) vs. normal BMI/T1]. Several inflammation markers were prospectively associated with endometrial cancer, including adipokines, pro- and anti-inflammatory cytokines, angiogenic factors and acute phase proteins. Inverse associations with anti-inflammatory markers (IL13, IL21), other inflammation markers/mediators (CCL3, IL1B, IL23), and a robust positive association between VEGFA and endometrial cancer risk were independent of BMI and estradiol, suggesting that these factors may influence risk through other mechanisms.
Collapse
Affiliation(s)
- Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Ronald C Eldridge
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Meredith S Shiels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Troy J Kemp
- HPV Immunology Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, MD
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec (CHUQ) Research Center, Laval University, Faculty of Pharmacy, Québec, Canada
| | - Patricia Hartge
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Mark E Sherman
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD
| | - Louise A Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Amanda Black
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Anil K Chaturvedi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Mahboobeh Safaeian
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Ligia Pinto
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
- HPV Immunology Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc, Frederick, MD
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| |
Collapse
|
29
|
Patel YM, Park SL, Han Y, Wilkens LR, Bickeböller H, Rosenberger A, Caporaso N, Landi MT, Brüske I, Risch A, Wei Y, Christiani DC, Brennan P, Houlston R, McKay J, McLaughlin J, Hung R, Murphy S, Stram DO, Amos C, Le Marchand L. Novel Association of Genetic Markers Affecting CYP2A6 Activity and Lung Cancer Risk. Cancer Res 2016; 76:5768-5776. [PMID: 27488534 PMCID: PMC5050097 DOI: 10.1158/0008-5472.can-16-0446] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 06/10/2016] [Indexed: 01/28/2023]
Abstract
Metabolism of nicotine by cytochrome P450 2A6 (CYP2A6) is a suspected determinant of smoking dose and, consequently, lung cancer risk. We conducted a genome-wide association study (GWAS) of CYP2A6 activity, as measured by the urinary ratio of trans-3'-hydroxycotinine and its glucuronide conjugate over cotinine (total 3HCOT/COT), among 2,239 smokers in the Multiethnic Cohort (MEC) study. We identified 248 CYP2A6 variants associated with CYP2A6 activity (P < 5 × 10-8). CYP2A6 activity was correlated (r = 0.32; P < 0.0001) with total nicotine equivalents (a measure of nicotine uptake). When we examined the effect of these variants on lung cancer risk in the Transdisciplinary Research in Cancer of the Lung (TRICL) consortium GWAS dataset (13,479 cases and 43,218 controls), we found that the vast majority of these individual effects were directionally consistent and associated with an increased lung cancer risk. Two hundred and twenty-six of the 248 variants associated with CYP2A6 activity in the MEC were available in TRICL. Of them, 81% had directionally consistent risk estimates, and six were globally significantly associated with lung cancer. When conditioning on nine known functional variants and two deletions, the top two SNPs (rs56113850 in MEC and rs35755165 in TRICL) remained significantly associated with CYP2A6 activity in MEC and lung cancer in TRICL. The present data support the hypothesis that a greater CYP2A6 activity causes smokers to smoke more extensively and be exposed to higher levels of carcinogens, resulting in an increased risk for lung cancer. Although the variants identified in these studies may be used as risk prediction markers, the exact causal variants remain to be identified. Cancer Res; 76(19); 5768-76. ©2016 AACR.
Collapse
Affiliation(s)
- Yesha M Patel
- Department of Preventive Medicine and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Sunghim L Park
- Department of Preventive Medicine and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Younghun Han
- Department of Biomedical Data Science, Dartmouth College, Hanover, New Hampshire
| | - Lynne R Wilkens
- Epidemiology Program, University of Hawai'i Cancer Center, Honolulu, Hawaii
| | - Heike Bickeböller
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| | - Albert Rosenberger
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| | - Neil Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Irene Brüske
- Helmholtz Centre Munich, German Research Centre for Environmental Health, Institute of Epidemiology I, Neuherberg, Germany
| | - Angela Risch
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Yongyue Wei
- Nanjing Medical University School of Public Health, Nanjing, China
| | - David C Christiani
- Massachusetts General Hospital, Boston, Massachusetts. Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts
| | - Paul Brennan
- Genetic Epidemiology Group, International Agency for Research on Cancer (IARC), Lyon, France
| | - Richard Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - James McKay
- Genetic Epidemiology Group, International Agency for Research on Cancer (IARC), Lyon, France
| | | | - Rayjean Hung
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Sharon Murphy
- Department of Biochemistry Molecular Biology and Biophysics and Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Daniel O Stram
- Department of Preventive Medicine and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Christopher Amos
- Department of Biomedical Data Science, Dartmouth College, Hanover, New Hampshire
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawai'i Cancer Center, Honolulu, Hawaii.
| |
Collapse
|
30
|
Serum metabolomic profiling of prostate cancer risk in the prostate, lung, colorectal, and ovarian cancer screening trial. Br J Cancer 2016; 115:1087-1095. [PMID: 27673363 PMCID: PMC5117796 DOI: 10.1038/bjc.2016.305] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/16/2016] [Accepted: 08/24/2016] [Indexed: 01/21/2023] Open
Abstract
Background: Two recent metabolomic analyses found serum lipid, energy, and other metabolites related to aggressive prostate cancer risk up to 20 years prior to diagnosis. Methods: We conducted a serum metabolomic investigation of prostate cancer risk in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial that included annual serum total prostate-specific antigen measurement and digital rectal examination. This nested study included 380 cases diagnosed post-screening and 380 controls individually matched to cases on age, race, study centre, and blood-collection date (median time to diagnosis, 10 years (range 4.4–17 years)). Sera were analysed on a high-resolution accurate mass platform of ultrahigh-performance liquid and gas chromatography/mass spectroscopy that identified 695 known metabolites. Logistic regression conditioned on the matching factors estimated odds ratios (OR) and 95% confidence intervals of risk associated with an 80th percentile increase in the log-metabolite signal. Results: Twenty-seven metabolites were associated with prostate cancer at P<0.05. Pyroglutamine, gamma-glutamylphenylalanine, phenylpyruvate, N-acetylcitrulline, and stearoylcarnitine showed the strongest metabolite-risk signals (ORs=0.53, 0.51, 0.46, 0.58, and 1.74, respectively; 0.001⩽P⩽0.006). Findings were similar for aggressive disease (peptide chemical class, P=0.03). None of the P-values were below the threshold of Bonferroni correction, however. Conclusions: A unique metabolomic profile associated with post-screening prostate cancer is identified that differs from that in a previously studied, unscreened population.
Collapse
|
31
|
Falk RT, Staff AC, Bradwin G, Karumanchi SA, Troisi R. A prospective study of angiogenic markers and postmenopausal breast cancer risk in the prostate, lung, colorectal, and ovarian cancer screening trial. Cancer Causes Control 2016; 27:1009-17. [PMID: 27357932 PMCID: PMC4958123 DOI: 10.1007/s10552-016-0779-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/11/2016] [Indexed: 11/12/2022]
Abstract
PURPOSE Pro-angiogenic factors are positively associated with breast tumor staging and poorer prognosis, but their role in the etiology of breast cancer has not been assessed. METHODS We measured serum levels of the pro-angiogenic vascular endothelial growth factor A (VEGF), and placental growth factor (PlGF) and anti-angiogenic soluble fms-like tyrosine kinase-1 (sFlt-1) in 352 incident breast cancer cases [mean age at diagnosis 67 (range 55-83)] and 352 non-cases in the prostate, lung, colorectal, and ovarian screening trial (women enrolled 1993-2001, followed through 2005) matched on age and date of enrollment. Cases were followed on average 4.2 years from blood draw to diagnosis, range 3.9-12.8 years; 53 % were estrogen receptor positive/progesterone receptor positive (ER+/PR+), and 13 % were ER-/PR-. Quartile-specific hazard ratios (HR) and 95 % confidence intervals (CI) were estimated using weighted Cox proportional hazards regression models adjusted for known breast cancer risk factors. An ordinal variable for the angiogenic markers was used to test for trend in the HR. RESULTS Comparing the highest to lowest quartile, multivariable HR were 0.90 for VEGF (95 % CI 0.33-2.43, p trend = 0.88), 1.38 for sFlt-1 (95 % CI 0.63-3.04, p trend = 0.63), and 0.62 for PlGF (95 % CI 0.19-2.00, p trend = 0.73). Risk patterns were not altered when all angiogenic markers were included in the model simultaneously, or by restricting analyses to invasive breast cancers, to cases diagnosed two or more years after blood collection or to ER+ tumors. CONCLUSIONS There was no evidence of an increased breast cancer risk associated with circulating levels of pro-angiogenic markers VEGF and PlGF or a reduced risk with circulating levels of anti-angiogenic marker sFlt-1.
Collapse
Affiliation(s)
- Roni T. Falk
- />Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, 9609 Medical Center Drive, Bethesda, MD 20852 USA
| | - Annetine Cathrine Staff
- />Women and Children’s Division, Department of Gynecology and Obstetrics, Oslo University Hospital, Ullevål, 0424 Oslo, Norway
| | - Gary Bradwin
- />Clinical and Epidemiologic Research Laboratory, Department of Laboratory Medicine, Boston Children’s Hospital, Boston, MA USA
| | - S. Ananth Karumanchi
- />Deparments of Medicine, Obstetrics and Gynecology, Center for Vascular Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA USA
| | - Rebecca Troisi
- />Epidemiology and Biostatistics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD 20852 USA
| |
Collapse
|
32
|
Carrick DM, Black A, Gohagan JK, Khan A, Pettit K, Williams C, Yu K, Yurgalevitch S, Huang WY, Zhu C. The PLCO Biorepository: Creating, Maintaining, and Administering a Unique Biospecimen Resource. Rev Recent Clin Trials 2016; 10:212-22. [PMID: 26238117 DOI: 10.2174/1574887110666150730121429] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 07/14/2015] [Accepted: 07/16/2015] [Indexed: 12/12/2022]
Abstract
Inclusion of biospecimens in population-based studies is an integral part of understanding disease etiology, identifying biomarkers and developing prevention and treatment strategies. The Prostate, Lung, Colorectal, and Ovarian (PLCO) cancer screening trial collected, processed and stored biospecimens from participants to create a biorepository of specimens which serves as a useful resource for a broad research community. PLCO collected blood samples from consented screening arm participants at six screening rounds and a buccal sample from consented control arm participants. In addition, formalin-fixed paraffin embedded tumor tissue specimens were collected for participants in both arms for selected cancer sites. Collection of biospecimens at multiple timepoints (i.e. serial samples) and prior to cancer diagnosis, paired with rich epidemiologic and screening data, makes the PLCO collection of biospecimens a uniquely valuable resource. As such, access to the PLCO biorepository is granted to investigators by a rigorous scientific review process and guided by a steering committee which is responsible for developing and implementing the biospecimen use policies. Here, we describe the procedures for biospecimen collection, processing, storage, requisition, and distribution, as well as data management employed in PLCO. We also provide examples of how the biospecimens have been used to advance cancer research and describe relevant lessons learned to help inform cohorts wishing to add or modify biospecimen collection.
Collapse
Affiliation(s)
- Danielle M Carrick
- Epidemiology and Genomics Research Program DCCPS, NCI, NIH, 9609 Medical Center Drive 4E224 Rockville, MD 20850, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Qian DC, Han Y, Byun J, Shin HR, Hung RJ, McLaughlin JR, Landi MT, Seminara D, Amos CI. A Novel Pathway-Based Approach Improves Lung Cancer Risk Prediction Using Germline Genetic Variations. Cancer Epidemiol Biomarkers Prev 2016; 25:1208-15. [PMID: 27222311 DOI: 10.1158/1055-9965.epi-15-1318] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 05/13/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Although genome-wide association studies (GWAS) have identified many genetic variants that are strongly associated with lung cancer, these variants have low penetrance and serve as poor predictors of lung cancer in individuals. We sought to increase the predictive value of germline variants by considering their cumulative effects in the context of biologic pathways. METHODS For individuals in the Environment and Genetics in Lung Cancer Etiology study (1,815 cases/1,971 controls), we computed pathway-level susceptibility effects as the sum of relevant SNP variant alleles weighted by their log-additive effects from a separate lung cancer GWAS meta-analysis (7,766 cases/37,482 controls). Logistic regression models based on age, sex, smoking, genetic variants, and principal components of pathway effects and pathway-smoking interactions were trained and optimized in cross-validation and further tested on an independent dataset (556 cases/830 controls). We assessed prediction performance using area under the receiver operating characteristic curve (AUC). RESULTS Compared with typical binomial prediction models that have epidemiologic predictors (AUC = 0.607) in addition to top GWAS variants (AUC = 0.617), our pathway-based smoking-interactive multinomial model significantly improved prediction performance in external validation (AUC = 0.656, P < 0.0001). CONCLUSIONS Our biologically informed approach demonstrated a larger increase in AUC over nongenetic counterpart models relative to previous approaches that incorporate variants. IMPACT This model is the first of its kind to evaluate lung cancer prediction using subtype-stratified genetic effects organized into pathways and interacted with smoking. We propose pathway-exposure interactions as a potentially powerful new contributor to risk inference. Cancer Epidemiol Biomarkers Prev; 25(8); 1208-15. ©2016 AACR.
Collapse
Affiliation(s)
- David C Qian
- Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire
| | - Younghun Han
- Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire
| | - Jinyoung Byun
- Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire
| | - Hae Ri Shin
- Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Canada
| | - John R McLaughlin
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | | | | | - Christopher I Amos
- Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire.
| |
Collapse
|
34
|
Agalliu I, Gapstur S, Chen Z, Wang T, Anderson RL, Teras L, Kreimer AR, Hayes RB, Freedman ND, Burk RD. Associations of Oral α-, β-, and γ-Human Papillomavirus Types With Risk of Incident Head and Neck Cancer. JAMA Oncol 2016; 2:599-606. [PMID: 26794505 DOI: 10.1001/jamaoncol.2015.5504] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Importance Prospective studies are needed to examine the temporal relationship between oral human papillomavirus (HPV) detection and risk of head and neck squamous cell carcinoma (HNSCC). Moreover, the oral cavity contains a wide spectrum of α-, β-, and γ-HPV types, but their association with risk of HNSCC is unknown. Objective To prospectively examine associations between α-, β-, and γ-HPV detection in the oral cavity and incident HNSCC. Design A nested case-control study was carried out among 96 650 participants, cancer free at baseline, with available mouthwash samples in 2 prospective cohort studies: (1) the American Cancer Society Cancer Prevention Study II Nutrition Cohort and (2) the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. Incident cases of HNSCC (n = 132) were identified during an average 3.9 years of follow-up in both cohorts. Three controls per case (n = 396) were selected through incidence density sampling and matched on age, sex, race/ethnicity, and time since mouthwash collection. Methods Through a next-generation sequencing assay, DNA from α-, β-, and γ-HPV types were detected. Conditional logistic regression models were used to estimate odds ratios (ORs) and 95% CIs, adjusting for smoking history, alcohol consumption, and detection of HPV-16 for β- and γ-HPVs. Main Outcomes and Measures Incident HNSCC, which includes cancers of the oropharynx, oral cavity, and larynx. Results A total of 132 participants developed HNSCC during the follow-up period (103 men and 29 women; average age at baseline, 66.5 years). Oral HPV-16 detection was associated with incident HNSCC (OR, 7.1; 95% CI, 2.2-22.6), with positive association for oropharyngeal SCC (OR, 22.4; 95% CI, 1.8-276.7), but not for oral cavity (OR, 4.5; 95% CI, 0.6-34.7) or laryngeal SCCs (OR, 0.11; 95% CI, 0.01-834.80). Detection of β1-HPV-5 and β2-HPV-38 types, as well as γ-11 and γ-12 species, had ORs for HNSCC that ranged from 2.64 to 5.45 (P < .01 for all comparisons). Detection of β1-HPV-5 type was associated with oropharyngeal (OR, 7.42; 95% CI, 0.98-56.82; P = .054), oral cavity (OR, 5.34; 95% CI, 1.51-18.80; P = .01), and laryngeal SCCs (OR, 2.71; 95% CI, 1.00-7.43; P = .05), whereas γ11- and γ12-HPV species were associated with both oral cavity (OR, 7.47; 95% CI, 1.21-46.17; P = .03; and OR, 6.71; 95% CI, 1.47-30.75; P = .01, respectively) and laryngeal SCCs (OR, 7.49; 95% CI, 1.10-51.04; P = .04 and OR, 5.31; 95% CI, 1.13-24.95; P = .03, respectively). Conclusions and Relevance This study demonstrates that HPV-16 detection precedes the incidence of oropharyngeal SCC. Associations of other HPVs, including γ11- and γ12-HPV species and β1-HPV-5 type suggest a broader role for HPVs in HNSCC etiology.
Collapse
Affiliation(s)
- Ilir Agalliu
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | | | - Zigui Chen
- Department of Pediatrics (Genetics), Albert Einstein College of Medicine, Bronx, New York
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | | | | | - Aimée R Kreimer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Richard B Hayes
- Department of Population Health and Environmental Medicine, New York University, New York
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Robert D Burk
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York3Department of Pediatrics (Genetics), Albert Einstein College of Medicine, Bronx, New York6Departments of Microbiology and Immunology and Obstetrics, Gyn
| |
Collapse
|
35
|
Qian DC, Byun J, Han Y, Greene CS, Field JK, Hung RJ, Brhane Y, Mclaughlin JR, Fehringer G, Landi MT, Rosenberger A, Bickeböller H, Malhotra J, Risch A, Heinrich J, Hunter DJ, Henderson BE, Haiman CA, Schumacher FR, Eeles RA, Easton DF, Seminara D, Amos CI. Identification of shared and unique susceptibility pathways among cancers of the lung, breast, and prostate from genome-wide association studies and tissue-specific protein interactions. Hum Mol Genet 2015; 24:7406-20. [PMID: 26483192 PMCID: PMC4664175 DOI: 10.1093/hmg/ddv440] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/11/2015] [Accepted: 10/12/2015] [Indexed: 12/18/2022] Open
Abstract
Results from genome-wide association studies (GWAS) have indicated that strong single-gene effects are the exception, not the rule, for most diseases. We assessed the joint effects of germline genetic variations through a pathway-based approach that considers the tissue-specific contexts of GWAS findings. From GWAS meta-analyses of lung cancer (12 160 cases/16 838 controls), breast cancer (15 748 cases/18 084 controls) and prostate cancer (14 160 cases/12 724 controls) in individuals of European ancestry, we determined the tissue-specific interaction networks of proteins expressed from genes that are likely to be affected by disease-associated variants. Reactome pathways exhibiting enrichment of proteins from each network were compared across the cancers. Our results show that pathways associated with all three cancers tend to be broad cellular processes required for growth and survival. Significant examples include the nerve growth factor (P = 7.86 × 10(-33)), epidermal growth factor (P = 1.18 × 10(-31)) and fibroblast growth factor (P = 2.47 × 10(-31)) signaling pathways. However, within these shared pathways, the genes that influence risk largely differ by cancer. Pathways found to be unique for a single cancer focus on more specific cellular functions, such as interleukin signaling in lung cancer (P = 1.69 × 10(-15)), apoptosis initiation by Bad in breast cancer (P = 3.14 × 10(-9)) and cellular responses to hypoxia in prostate cancer (P = 2.14 × 10(-9)). We present the largest comparative cross-cancer pathway analysis of GWAS to date. Our approach can also be applied to the study of inherited mechanisms underlying risk across multiple diseases in general.
Collapse
Affiliation(s)
- David C Qian
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Jinyoung Byun
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Younghun Han
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Casey S Greene
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John K Field
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool Cancer Research Centre, Liverpool L69 3GA, UK
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Yonathan Brhane
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - John R Mclaughlin
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Gordon Fehringer
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Maria Teresa Landi
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Albert Rosenberger
- Department of Genetic Epidemiology, University Medical Centre Göttingen, 37099 Göttingen, Germany
| | - Heike Bickeböller
- Department of Genetic Epidemiology, University Medical Centre Göttingen, 37099 Göttingen, Germany
| | - Jyoti Malhotra
- Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Angela Risch
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Joachim Heinrich
- Institute of Epidemiology I, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - David J Hunter
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Brian E Henderson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Fredrick R Schumacher
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Rosalind A Eeles
- Department of Cancer Genetics, Institute of Cancer Research, London SW7 3RP, UK and
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge CB1 8RN, UK
| | - Daniela Seminara
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christopher I Amos
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA,
| |
Collapse
|
36
|
Loftfield E, Freedman ND, Graubard BI, Guertin KA, Black A, Huang WY, Shebl FM, Mayne ST, Sinha R. Association of Coffee Consumption With Overall and Cause-Specific Mortality in a Large US Prospective Cohort Study. Am J Epidemiol 2015; 182:1010-22. [PMID: 26614599 PMCID: PMC5875735 DOI: 10.1093/aje/kwv146] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 06/01/2015] [Indexed: 01/06/2023] Open
Abstract
Concerns about high caffeine intake and coffee as a vehicle for added fat and sugar have raised questions about the net impact of coffee on health. Although inverse associations have been observed for overall mortality, data for cause-specific mortality are sparse. Additionally, few studies have considered exclusively decaffeinated coffee intake or use of coffee additives. Coffee intake was assessed at baseline by self-report in the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial. Hazard ratios were estimated using Cox proportional hazards models. Among 90,317 US adults without cancer at study baseline (1998-2001) or history of cardiovascular disease at study enrollment (1993-2001), 8,718 deaths occurred during 805,644 person-years of follow-up from 1998 through 2009. Following adjustment for smoking and other potential confounders, coffee drinkers, as compared with nondrinkers, had lower hazard ratios for overall mortality (<1 cup/day: hazard ratio (HR) = 0.99 (95% confidence interval (CI): 0.92, 1.07); 1 cup/day: HR = 0.94 (95% CI: 0.87, 1.02); 2-3 cups/day: HR = 0.82 (95% CI: 0.77, 0.88); 4-5 cups/day: HR = 0.79 (95% CI: 0.72, 0.86); ≥6 cups/day: HR = 0.84 (95% CI: 0.75, 0.95)). Similar findings were observed for decaffeinated coffee and coffee additives. Inverse associations were observed for deaths from heart disease, chronic respiratory diseases, diabetes, pneumonia and influenza, and intentional self-harm, but not cancer. Coffee may reduce mortality risk by favorably affecting inflammation, lung function, insulin sensitivity, and depression.
Collapse
Affiliation(s)
- Erikka Loftfield
- Correspondence to Dr. Erikka Loftfield, Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI Shady Grove, National Cancer Institute, 9609 Medical Center Drive 6E332, Rockville, MD 20850-9768 (e-mail: )
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Brenner DR, Amos CI, Brhane Y, Timofeeva MN, Caporaso N, Wang Y, Christiani DC, Bickeböller H, Yang P, Albanes D, Stevens VL, Gapstur S, McKay J, Boffetta P, Zaridze D, Szeszenia-Dabrowska N, Lissowska J, Rudnai P, Fabianova E, Mates D, Bencko V, Foretova L, Janout V, Krokan HE, Skorpen F, Gabrielsen ME, Vatten L, Njølstad I, Chen C, Goodman G, Lathrop M, Vooder T, Välk K, Nelis M, Metspalu A, Broderick P, Eisen T, Wu X, Zhang D, Chen W, Spitz MR, Wei Y, Su L, Xie D, She J, Matsuo K, Matsuda F, Ito H, Risch A, Heinrich J, Rosenberger A, Muley T, Dienemann H, Field JK, Raji O, Chen Y, Gosney J, Liloglou T, Davies MPA, Marcus M, McLaughlin J, Orlow I, Han Y, Li Y, Zong X, Johansson M, Liu G, Tworoger SS, Le Marchand L, Henderson BE, Wilkens LR, Dai J, Shen H, Houlston RS, Landi MT, Brennan P, Hung RJ. Identification of lung cancer histology-specific variants applying Bayesian framework variant prioritization approaches within the TRICL and ILCCO consortia. Carcinogenesis 2015; 36:1314-26. [PMID: 26363033 PMCID: PMC4635669 DOI: 10.1093/carcin/bgv128] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 08/17/2015] [Accepted: 08/24/2015] [Indexed: 01/08/2023] Open
Abstract
Large-scale genome-wide association studies (GWAS) have likely uncovered all common variants at the GWAS significance level. Additional variants within the suggestive range (0.0001> P > 5×10(-8)) are, however, still of interest for identifying causal associations. This analysis aimed to apply novel variant prioritization approaches to identify additional lung cancer variants that may not reach the GWAS level. Effects were combined across studies with a total of 33456 controls and 6756 adenocarcinoma (AC; 13 studies), 5061 squamous cell carcinoma (SCC; 12 studies) and 2216 small cell lung cancer cases (9 studies). Based on prior information such as variant physical properties and functional significance, we applied stratified false discovery rates, hierarchical modeling and Bayesian false discovery probabilities for variant prioritization. We conducted a fine mapping analysis as validation of our methods by examining top-ranking novel variants in six independent populations with a total of 3128 cases and 2966 controls. Three novel loci in the suggestive range were identified based on our Bayesian framework analyses: KCNIP4 at 4p15.2 (rs6448050, P = 4.6×10(-7)) and MTMR2 at 11q21 (rs10501831, P = 3.1×10(-6)) with SCC, as well as GAREM at 18q12.1 (rs11662168, P = 3.4×10(-7)) with AC. Use of our prioritization methods validated two of the top three loci associated with SCC (P = 1.05×10(-4) for KCNIP4, represented by rs9799795) and AC (P = 2.16×10(-4) for GAREM, represented by rs3786309) in the independent fine mapping populations. This study highlights the utility of using prior functional data for sequence variants in prioritization analyses to search for robust signals in the suggestive range.
Collapse
Affiliation(s)
- Darren R Brenner
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Ontario M5T 3L9, Canada, Section of Genetics, International Agency for Research on Cancer, 69372 Lyon, France, Department of Cancer Epidemiology and Prevention Research, Cancer Control Alberta, Alberta Health Services, Calgary, Alberta T2T 5C7, Canada
| | - Christopher I Amos
- Department of Community and Family Medicine, Center for Genomic Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03766, USA
| | - Yonathan Brhane
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Ontario M5T 3L9, Canada
| | - Maria N Timofeeva
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH8 9YL, UK
| | - Neil Caporaso
- Department of Health and Human Services, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yufei Wang
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | - David C Christiani
- Departments of Environmental Health and Epidemiology, Harvard University School of Public Health, Boston, MA 02115, USA
| | - Heike Bickeböller
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Ping Yang
- Division of Health Sciences, Cancer Center and College of Medicine, Mayo Clinic, Rochester, NY 55905, USA
| | - Demetrius Albanes
- Department of Health and Human Services, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Victoria L Stevens
- Epidemiology Research Program, American Cancer Society, Epidemiology and Surveillance Research, Atlanta, GA 30301, USA
| | - Susan Gapstur
- Epidemiology Research Program, American Cancer Society, Epidemiology and Surveillance Research, Atlanta, GA 30301, USA
| | - James McKay
- Section of Genetics, International Agency for Research on Cancer, 69372 Lyon, France
| | - Paolo Boffetta
- Population Sciences, Tisch Cancer Center and Institute for Translational Epidemiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David Zaridze
- Institute of Carcinogenesis, Russian N.N.Blokhin Cancer Research Centre, 115478 Moscow, Russia
| | | | - Jolanta Lissowska
- Department of Epidemiology and Cancer Prevention, The M. Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw 02781, Poland
| | - Peter Rudnai
- National Institute of Environmental Health, Budapest 1097, Hungary
| | - Eleonora Fabianova
- Department of Health Risk Assessment, Regional Authority of Public Health, Banská Bystrica 97556, Slovak Republic
| | - Dana Mates
- National Institute of Public Health, Bucharest 050463, Romania
| | - Vladimir Bencko
- Institute of Hygiene and Epidemiology, 1st Faculty of Medicine, Charles University in Prague, 128 00 Prague 2, Czech Republic
| | - Lenka Foretova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Brno 65653, Czech Republic
| | - Vladimir Janout
- Department of Preventive Medicine, Palacky University, Olomouc 77515, Czech Republic
| | - Hans E Krokan
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine
| | - Frank Skorpen
- Department of Laboratory Medicine, Children's and Women's Health, Faculty of Medicine and
| | - Maiken E Gabrielsen
- Department of Laboratory Medicine, Children's and Women's Health, Faculty of Medicine and
| | - Lars Vatten
- Department of Public Health and General Practice, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim 7489, Norway
| | - Inger Njølstad
- Department of Community Medicine, University of Tromso, Tromso N-9037, Norway
| | - Chu Chen
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Gary Goodman
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Mark Lathrop
- McGill University and Genome Québec Innovation Centre, Montréal, Quebec, Canada
| | - Tõnu Vooder
- Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia
| | - Kristjan Välk
- Department of Biomedicine, University of Bergen, Bergen 5009, Norway
| | - Mari Nelis
- Institute of Molecular and Cell Biology, Estonian Biocentre, Genotyping Core Facility, Tartu 51010, Estonia
| | - Andres Metspalu
- Institute of Molecular and Cell Biology, Estonian Biocentre, Genotyping Core Facility, Tartu 51010, Estonia
| | - Peter Broderick
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | - Timothy Eisen
- Department of Oncology, Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| | - Xifeng Wu
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Di Zhang
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wei Chen
- Department of Genetics, U.T. M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Margaret R Spitz
- Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yongyue Wei
- Departments of Environmental Health and Epidemiology, Harvard University School of Public Health, Boston, MA 02115, USA
| | - Li Su
- Departments of Environmental Health and Epidemiology, Harvard University School of Public Health, Boston, MA 02115, USA
| | - Dong Xie
- Division of Health Sciences, Cancer Center and College of Medicine, Mayo Clinic, Rochester, NY 55905, USA
| | - Jun She
- Division of Health Sciences, Cancer Center and College of Medicine, Mayo Clinic, Rochester, NY 55905, USA
| | - Keitaro Matsuo
- Department of Preventive Medicine, Kyushu University Graduate School of Medicine, Fukuoka City 819-0395, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Hidemi Ito
- Department of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Chikusa-ku Nagoya 464-0021, Japan
| | - Angela Risch
- Division of Epigenomics and Cancer Risk Factors, DKFZ, 69121 Heidelberg, Germany, Division of Epigenomics and Cancer Risk Factors, Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), 69121 Heidelberg, Germany
| | - Joachim Heinrich
- Unit of Environmental Epidemiology, Helmholtz Zentrum Munchen, 85764 Neuherberg, Germany
| | - Albert Rosenberger
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Thomas Muley
- Division of Epigenomics and Cancer Risk Factors, Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), 69121 Heidelberg, Germany, Translational Research Unit and
| | - Hendrik Dienemann
- Division of Epigenomics and Cancer Risk Factors, Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), 69121 Heidelberg, Germany, Department of Thoracic Surgery, Thoraxklinik am Universitätsklinikum Heidelberg, 69117 Heidelberg, Germany
| | - John K Field
- Roy Castle Lung Cancer Research Programme, The University of Liverpool Cancer Research Centre, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, The University of Liverpool, Liverpool L69 3BX, UK
| | - Olaide Raji
- Roy Castle Lung Cancer Research Programme, The University of Liverpool Cancer Research Centre, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, The University of Liverpool, Liverpool L69 3BX, UK
| | - Ying Chen
- Roy Castle Lung Cancer Research Programme, The University of Liverpool Cancer Research Centre, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, The University of Liverpool, Liverpool L69 3BX, UK
| | - John Gosney
- Roy Castle Lung Cancer Research Programme, The University of Liverpool Cancer Research Centre, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, The University of Liverpool, Liverpool L69 3BX, UK
| | - Triantafillos Liloglou
- Roy Castle Lung Cancer Research Programme, The University of Liverpool Cancer Research Centre, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, The University of Liverpool, Liverpool L69 3BX, UK
| | - Michael P A Davies
- Roy Castle Lung Cancer Research Programme, The University of Liverpool Cancer Research Centre, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, The University of Liverpool, Liverpool L69 3BX, UK
| | - Michael Marcus
- Roy Castle Lung Cancer Research Programme, The University of Liverpool Cancer Research Centre, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, The University of Liverpool, Liverpool L69 3BX, UK
| | - John McLaughlin
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Ontario M5T 3L9, Canada
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Younghun Han
- Department of Community and Family Medicine, Center for Genomic Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03766, USA
| | - Yafang Li
- Department of Community and Family Medicine, Center for Genomic Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03766, USA
| | - Xuchen Zong
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Ontario M5T 3L9, Canada
| | - Mattias Johansson
- Section of Genetics, International Agency for Research on Cancer, 69372 Lyon, France
| | - Geoffrey Liu
- Medical Oncology and Haematology, Department of Medicine, Princess Margaret Hospital, Toronto, Ontario M5G 2M9, Canada
| | - Shelley S Tworoger
- Department of Epidemiology, Harvard School of Public Health, Boston, MA 02115, USA, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Loic Le Marchand
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Brian E Henderson
- Keck School of Medicine, University of South California, Los Angeles, CA 90089-0911, USA and
| | - Lynne R Wilkens
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Juncheng Dai
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | - Maria T Landi
- Department of Health and Human Services, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paul Brennan
- Section of Genetics, International Agency for Research on Cancer, 69372 Lyon, France
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Ontario M5T 3L9, Canada,
| |
Collapse
|
38
|
Barry KH, Moore LE, Liao LM, Huang WY, Andreotti G, Poulin M, Berndt SI. Prospective study of DNA methylation at LINE-1 and Alu in peripheral blood and the risk of prostate cancer. Prostate 2015; 75:1718-25. [PMID: 26250474 PMCID: PMC4535169 DOI: 10.1002/pros.23053] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 07/02/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Evidence suggests that global blood DNA methylation levels may be associated with the risk of various cancers, but no studies have evaluated this relationship for prostate cancer. METHODS We used pyrosequencing to quantify DNA methylation levels at the long interspersed nuclear element 1 (LINE-1) and Alu repetitive elements in pre-diagnostic blood samples from 694 prostate cancer cases and 703 controls from the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. We evaluated prostate cancer risk associated with the mean methylation level for each element using logistic regression, adjusting for potential confounders. RESULTS We did not observe a significant association with prostate cancer for LINE-1 [odds ratio (OR) for the highest compared to the lowest quartile = 1.01, 95% confidence interval (CI): 0.73-1.39, Ptrend = 0.99] or Alu (OR = 0.94, 95% CI: 0.68-1.29, Ptrend = 0.69) methylation levels overall. However, for Alu, we observed that higher DNA methylation levels were associated with a significant increased risk for those diagnosed 4 or more years after blood draw (OR = 2.26, 95% CI: 1.27-4.00, Ptrend = 4.4 × 10(-3) ). In contrast, there was no association for those diagnosed 2 (OR = 1.13, 95% CI: 0.67-1.90, Ptrend = 0.64) or 3 years after draw (OR = 1.22, 95% CI: 0.71-2.07, Ptrend = 0.32), and a decreased risk for those diagnosed less than 2 years after draw (OR = 0.40, 95% CI: 0.25-0.65, Ptrend = 3.8 × 10(-5) ; Pheterogeneity = 5.3 × 10(-6) ). CONCLUSIONS Although LINE-1 DNA methylation levels were not associated with prostate cancer, we observed an association for Alu that varied by time from blood draw to diagnosis. Our study suggests that elevated Alu blood DNA methylation levels several years before diagnosis may be associated with an increased prostate cancer risk.
Collapse
Affiliation(s)
- Kathryn Hughes Barry
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Lee E. Moore
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Linda M. Liao
- Nutritional Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Wen-Yi Huang
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | - Gabriella Andreotti
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| | | | - Sonja I. Berndt
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD
| |
Collapse
|
39
|
Lavender N, Hein DW, Brock G, Kidd LCR. Evaluation of Oxidative Stress Response Related Genetic Variants, Pro-oxidants, Antioxidants and Prostate Cancer. AIMS MEDICAL SCIENCE 2015; 2:271-294. [PMID: 26636131 PMCID: PMC4664461 DOI: 10.3934/medsci.2015.4.271] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Oxidative stress and detoxification mechanisms have been commonly studied in Prostate Cancer (PCa) due to their function in the detoxification of potentially damaging reactive oxygen species (ROS) and carcinogens. However, findings have been either inconsistent or inconclusive. These mixed findings may, in part, relate to failure to consider interactions among oxidative stress response related genetic variants along with pro- and antioxidant factors. Methods We examined the effects of 33 genetic and 26 environmental oxidative stress and defense factors on PCa risk and disease aggressiveness among 2,286 men from the Cancer Genetic Markers of Susceptibility project (1,175 cases, 1,111 controls). Single and joint effects were analyzed using a comprehensive statistical approach involving logistic regression, multi-dimensionality reduction, and entropy graphs. Results Inheritance of one CYP2C8 rs7909236 T or two SOD2 rs2758331 A alleles was linked to a 1.3- and 1.4-fold increase in risk of developing PCa, respectively (p-value = 0.006–0.013). Carriers of CYP1B1 rs1800440GG, CYP2C8 rs1058932TC and, NAT2 (rs1208GG, rs1390358CC, rs7832071TT) genotypes were associated with a 1.3 to 2.2-fold increase in aggressive PCa [p-value = 0.04–0.001, FDR 0.088–0.939]. We observed a 23% reduction in aggressive disease linked to inheritance of one or more NAT2 rs4646247 A alleles (p = 0.04, FDR = 0.405). Only three NAT2 sequence variants remained significant after adjusting for multiple hypotheses testing, namely NAT2 rs1208, rs1390358, and rs7832071. Lastly, there were no significant gene-environment or gene-gene interactions associated with PCa outcomes. Conclusions Variations in genes involved in oxidative stress and defense pathways may modify PCa. Our findings do not firmly support the role of oxidative stress genetic variants combined with lifestyle/environmental factors as modifiers of PCa and disease progression. However, additional multi-center studies poised to pool genetic and environmental data are needed to make strong conclusions.
Collapse
Affiliation(s)
- Nicole Lavender
- Department of Pharmacology and Toxicology and James Graham Brown Cancer Center, University of Louisville, Louisville, KY
| | - David W Hein
- Department of Pharmacology and Toxicology and James Graham Brown Cancer Center, University of Louisville, Louisville, KY
| | - Guy Brock
- Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, KY
| | - La Creis R Kidd
- Department of Pharmacology and Toxicology and James Graham Brown Cancer Center, University of Louisville, Louisville, KY
| |
Collapse
|
40
|
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: 4.5] [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
|
41
|
Han SS, Rosenberg PS, Ghosh A, Landi MT, Caporaso NE, Chatterjee N. An exposure-weighted score test for genetic associations integrating environmental risk factors. Biometrics 2015; 71:596-605. [PMID: 26134142 DOI: 10.1111/biom.12328] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 02/01/2015] [Accepted: 03/01/2015] [Indexed: 11/30/2022]
Abstract
Current methods for detecting genetic associations lack full consideration of the background effects of environmental exposures. Recently proposed methods to account for environmental exposures have focused on logistic regressions with gene-environment interactions. In this report, we developed a test for genetic association, encompassing a broad range of risk models, including linear, logistic and probit, for specifying joint effects of genetic and environmental exposures. We obtained the test statistics by maximizing over a class of score tests, each of which involves modified standard tests of genetic association through a weight function. This weight function reflects the potential heterogeneity of the genetic effects by levels of environmental exposures under a particular model. Simulation studies demonstrate the robust power of these methods for detecting genetic associations under a wide range of scenarios. Applications of these methods are further illustrated using data from genome-wide association studies of type 2 diabetes with body mass index and of lung cancer risk with smoking.
Collapse
Affiliation(s)
- Summer S Han
- Department of Radiology, Stanford University School of Medicine, Palo Alto, California 94305, U.S.A
| | - Philip S Rosenberg
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, 9609 Medical Center Drive Suite, Rockville, Bethesda, Maryland 20852, U.S.A
| | - Arpita Ghosh
- Public Health Foundation of India, Vasant Kunj, New Delhi 110070, India
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, 9609 Medical Center Drive Suite, Rockville, Bethesda, Maryland 20852, U.S.A
| | - Neil E Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, 9609 Medical Center Drive Suite, Rockville, Bethesda, Maryland 20852, U.S.A
| | - Nilanjan Chatterjee
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, 9609 Medical Center Drive Suite, Rockville, Bethesda, Maryland 20852, U.S.A
| |
Collapse
|
42
|
Piper MR, Freedman DM, Robien K, Kopp W, Rager H, Horst RL, Stolzenberg-Solomon RZ. Vitamin D-binding protein and pancreatic cancer: a nested case-control study. Am J Clin Nutr 2015; 101:1206-15. [PMID: 25904602 PMCID: PMC4441803 DOI: 10.3945/ajcn.114.096016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 03/16/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Vitamin D-binding protein (DBP) is the primary carrier of 25-hydroxyvitamin D [25(OH)D] in the circulation. One prospective study in male smokers found a protective association between DBP and pancreatic cancer, particularly among men with higher 25(OH)D concentrations. OBJECTIVE The objective was to examine the association between DBP and pancreatic cancer risk in an American population. DESIGN We conducted a nested case-control study in the Prostate, Lung, Colorectal, and Ovarian Cancer screening trial cohort of men and women aged 55-74 y at baseline. Between 1993 and 2010, 295 incident pancreatic adenocarcinoma cases were reported (follow-up to 15.1 y). Two controls (n = 590) were matched to each case by age, race, sex, and month of blood draw. We calculated smoking- and diabetes-adjusted ORs and 95% CIs with the use of conditional logistic regression. RESULTS DBP concentration was not significantly associated with pancreatic cancer overall [highest (≥7149.4 nmol/L) vs. lowest (<3670.4 nmol/L) quintile; OR: 1.75; 95% CI: 0.91, 3.37; P-trend = 0.25]. For serum 25(OH)D compared with the referent (50 to <75 nmol/L), individuals in the highest group had a significantly higher risk (≥100 nmol/L; OR: 3.23; 95% CI: 1.24, 8.44), whereas those in the lowest group had no significant association (<25 nmol/L; OR: 2.50; 95% CI: 0.92, 6.81). Further adjustment for DBP did not alter this association. CONCLUSION Our results do not support the hypothesis that serum DBP or 25(OH)D plays a protective role in pancreatic cancer. This trial was registered at clinicaltrials.gov as NCT00339495.
Collapse
Affiliation(s)
- Marina R Piper
- From the Nutritional Epidemiology Branch (MRP and RZS-S) and the Radiation Epidemiology Branch (DMF), Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD; the Departments of Epidemiology and Biostatistics and Exercise Science, Milken Institute School of Public Health, George Washington University, Washington, DC (KR); the Clinical Support Laboratory, Leidos Biomedical Research Inc./Frederick National Laboratory for Cancer Research, Frederick, MD (WK and HR); and Heartland Assays Inc., Iowa State University, Ames, IA (RLH)
| | - D Michal Freedman
- From the Nutritional Epidemiology Branch (MRP and RZS-S) and the Radiation Epidemiology Branch (DMF), Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD; the Departments of Epidemiology and Biostatistics and Exercise Science, Milken Institute School of Public Health, George Washington University, Washington, DC (KR); the Clinical Support Laboratory, Leidos Biomedical Research Inc./Frederick National Laboratory for Cancer Research, Frederick, MD (WK and HR); and Heartland Assays Inc., Iowa State University, Ames, IA (RLH)
| | - Kim Robien
- From the Nutritional Epidemiology Branch (MRP and RZS-S) and the Radiation Epidemiology Branch (DMF), Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD; the Departments of Epidemiology and Biostatistics and Exercise Science, Milken Institute School of Public Health, George Washington University, Washington, DC (KR); the Clinical Support Laboratory, Leidos Biomedical Research Inc./Frederick National Laboratory for Cancer Research, Frederick, MD (WK and HR); and Heartland Assays Inc., Iowa State University, Ames, IA (RLH)
| | - William Kopp
- From the Nutritional Epidemiology Branch (MRP and RZS-S) and the Radiation Epidemiology Branch (DMF), Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD; the Departments of Epidemiology and Biostatistics and Exercise Science, Milken Institute School of Public Health, George Washington University, Washington, DC (KR); the Clinical Support Laboratory, Leidos Biomedical Research Inc./Frederick National Laboratory for Cancer Research, Frederick, MD (WK and HR); and Heartland Assays Inc., Iowa State University, Ames, IA (RLH)
| | - Helen Rager
- From the Nutritional Epidemiology Branch (MRP and RZS-S) and the Radiation Epidemiology Branch (DMF), Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD; the Departments of Epidemiology and Biostatistics and Exercise Science, Milken Institute School of Public Health, George Washington University, Washington, DC (KR); the Clinical Support Laboratory, Leidos Biomedical Research Inc./Frederick National Laboratory for Cancer Research, Frederick, MD (WK and HR); and Heartland Assays Inc., Iowa State University, Ames, IA (RLH)
| | - Ronald L Horst
- From the Nutritional Epidemiology Branch (MRP and RZS-S) and the Radiation Epidemiology Branch (DMF), Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD; the Departments of Epidemiology and Biostatistics and Exercise Science, Milken Institute School of Public Health, George Washington University, Washington, DC (KR); the Clinical Support Laboratory, Leidos Biomedical Research Inc./Frederick National Laboratory for Cancer Research, Frederick, MD (WK and HR); and Heartland Assays Inc., Iowa State University, Ames, IA (RLH)
| | - Rachael Z Stolzenberg-Solomon
- From the Nutritional Epidemiology Branch (MRP and RZS-S) and the Radiation Epidemiology Branch (DMF), Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD; the Departments of Epidemiology and Biostatistics and Exercise Science, Milken Institute School of Public Health, George Washington University, Washington, DC (KR); the Clinical Support Laboratory, Leidos Biomedical Research Inc./Frederick National Laboratory for Cancer Research, Frederick, MD (WK and HR); and Heartland Assays Inc., Iowa State University, Ames, IA (RLH).
| |
Collapse
|
43
|
Loftfield E, Shiels MS, Graubard BI, Katki HA, Chaturvedi AK, Trabert B, Pinto LA, Kemp TJ, Shebl FM, Mayne ST, Wentzensen N, Purdue MP, Hildesheim A, Sinha R, Freedman ND. Associations of Coffee Drinking with Systemic Immune and Inflammatory Markers. Cancer Epidemiol Biomarkers Prev 2015; 24:1052-60. [PMID: 25999212 DOI: 10.1158/1055-9965.epi-15-0038-t] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 04/28/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Coffee drinking has been inversely associated with mortality as well as cancers of the endometrium, colon, skin, prostate, and liver. Improved insulin sensitivity and reduced inflammation are among the hypothesized mechanisms by which coffee drinking may affect cancer risk; however, associations between coffee drinking and systemic levels of immune and inflammatory markers have not been well characterized. METHODS We used Luminex bead-based assays to measure serum levels of 77 immune and inflammatory markers in 1,728 older non-Hispanic Whites. Usual coffee intake was self-reported using a food frequency questionnaire. We used weighted multivariable logistic regression models to examine associations between coffee and dichotomized marker levels. We conducted statistical trend tests by modeling the median value of each coffee category and applied a 20% false discovery rate criterion to P values. RESULTS Ten of the 77 markers were nominally associated (P trend < 0.05) with coffee drinking. Five markers withstood correction for multiple comparisons and included aspects of the host response namely chemotaxis of monocytes/macrophages (IFNγ, CX3CL1/fractalkine, CCL4/MIP-1β), proinflammatory cytokines (sTNFRII), and regulators of cell growth (FGF-2). Heavy coffee drinkers had lower circulating levels of IFNγ [odds ratios (OR), 0.35; 95% confidence intervals (CI), 0.16-0.75], CX3CL1/fractalkine (OR, 0.25; 95% CI, 0.10-0.64), CCL4/MIP-1β (OR, 0.48; 95% CI, 0.24-0.99), FGF-2 (OR, 0.62; 95% CI, 0.28-1.38), and sTNFRII (OR, 0.34; 95% CI, 0.15-0.79) than non-coffee drinkers. CONCLUSIONS Lower circulating levels of inflammatory markers among coffee drinkers may partially mediate previously observed associations of coffee with cancer and other chronic diseases. IMPACT Validation studies, ideally controlled feeding trials, are needed to confirm these associations.
Collapse
Affiliation(s)
- Erikka Loftfield
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland. Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut.
| | - Meredith S Shiels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Barry I Graubard
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Hormuzd A Katki
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Anil K Chaturvedi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Ligia A Pinto
- HPV Immunology Laboratory, Leidos Biomedical Research, Inc., SAIC-Frederick/NCI-Frederick, Frederick, Maryland
| | - Troy J Kemp
- HPV Immunology Laboratory, Leidos Biomedical Research, Inc., SAIC-Frederick/NCI-Frederick, Frederick, Maryland
| | - Fatma M Shebl
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut. Yale Cancer Center, New Haven, Connecticut
| | - Susan T Mayne
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut. Yale Cancer Center, New Haven, Connecticut
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Mark P Purdue
- Ontario Institute for Cancer Research, Toronto, Canada
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Rashmi Sinha
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
44
|
Karami S, Andreotti G, Liao LM, Pfeiffer RM, Weinstein SJ, Purdue MP, Hofmann JN, Albanes D, Mannisto S, Moore LE. LINE1 methylation levels in pre-diagnostic leukocyte DNA and future renal cell carcinoma risk. Epigenetics 2015; 10:282-92. [PMID: 25647181 DOI: 10.1080/15592294.2015.1006505] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Higher levels of LINE1 methylation in blood DNA have been associated with increased kidney cancer risk using post-diagnostically collected samples; however, this association has never been examined using pre-diagnostic samples. We examined the association between LINE1 %5mC and renal cell carcinoma (RCC) risk using pre-diagnostic blood DNA from the United States-based, Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO) (215 cases/436 controls), and the Alpha-tocopherol, Beta-carotene Cancer Prevention Study (ATBC) of Finnish male smokers (191 cases/575 controls). Logistic regression adjusted for age at blood draw, study center, pack-years of smoking, body mass index, hypertension, dietary alcohol intake, family history of cancer, and sex was used to calculate odds ratios (ORs) and 95% confidence intervals (CIs) using cohort and sex-specific methylation categories. In PLCO, higher, although non-significant, RCC risk was observed for participants at or above median methylation level (M2) compared to those below the median (M1) (OR: 1.37, 95% CI: 0.96-1.95). The association was stronger in males (M2 vs. M1, OR: 1.54, 95% CI: 1.00-2.39) and statistically significant among male smokers (M2 vs. M1, OR: 2.60, 95% CI: 1.46-4.63). A significant interaction for smoking was also detected (P-interaction: 0.01). No association was found among females or female smokers. Findings for male smokers were replicated in ATBC (M2 vs. M1, OR: 1.31, 95% CI: 1.07-1.60). In a pooled analysis of PLCO and ATBC male smokers (281 cases/755 controls), the OR among subjects at or above median methylation level (M2) compared to those below the median (M1) was 1.89 (95% CI: 1.34-2.67, P-value: 3 x 10(-4)); a trend was also observed by methylation quartile (P-trend: 0.002). These findings suggest that higher LINE1 methylation levels measured prior to cancer diagnosis may be a biomarker of future RCC risk among male smokers.
Collapse
Affiliation(s)
- Sara Karami
- a Division of Cancer Epidemiology and Genetics (DCEG); US National Cancer Institute (NCI); National Institutes of Health (NIH); Department of Health and Human Services (DHHS) ; Rockville , MD USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Murphy N, Cross AJ, Huang WY, Rajabzadeh-Heshejin V, Stanczyk F, Hayes R, Gunter MJ. A prospective evaluation of C-peptide levels and colorectal adenoma incidence. Cancer Epidemiol 2015; 39:160-5. [PMID: 25592235 DOI: 10.1016/j.canep.2014.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 12/20/2014] [Accepted: 12/22/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Obesity is a recognised positive risk factor for colorectal adenoma and colorectal cancer. Obesity is associated with insulin resistance and compensatory hyperinsulinaemia, and circulating insulin and C-peptide, a biomarker of insulin levels, have been positively associated with colorectal cancer risk. However, whether a similar relationship exists for colorectal adenomas, an established colorectal cancer precursor, is unclear. METHODS In a nested case-control study of 273 colorectal adenoma cases and 355 matched controls from the screening arm of the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial, serum C-peptide levels were measured by a chemiluminescent immunometric assay. Multivariable unconditional logistic regression models were used to calculate odds ratios (OR) and 95% confidence intervals (CI) for colorectal adenoma within quartiles of C-peptide. Further, to explore the temporal stability of C-peptide, repeat samples from the incident adenoma cases (n=50) and controls (n=30), over a 5-year period were assayed and the intra-class correlations (ICC) estimated. RESULTS In a multivariable model that included established colorectal adenoma risk factors, C-peptide levels were not associated with colorectal adenoma (Q4 vs. Q1, OR 0.83, 95% CI: 0.52-1.31; P-trend 0.32); similar null associations were observed by gender, by adenoma subsite and for advanced adenomas. Among control participants, the ICC value over a 5-year period was 0.66. CONCLUSION Our results suggest that higher C-peptide levels were not associated with colorectal adenoma incidence in this study population. Other biological pathways associated with obesity may be more relevant to the early stages of colorectal tumorigenesis.
Collapse
Affiliation(s)
- Neil Murphy
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
| | - Amanda J Cross
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Vian Rajabzadeh-Heshejin
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Frank Stanczyk
- Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA, USA; Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Richard Hayes
- Division of Epidemiology, Department of Environmental Medicine, NYU Langone Medical Center, NYU Cancer Institute, New York, NY, USA
| | - Marc J Gunter
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| |
Collapse
|
46
|
Barry KH, Moore LE, Sampson J, Yan L, Meyer A, Oler AJ, Chung CC, Wang Z, Yeager M, Amundadottir L, Berndt SI. DNA methylation levels at chromosome 8q24 in peripheral blood are associated with 8q24 cancer susceptibility loci. Cancer Prev Res (Phila) 2014; 7:1282-92. [PMID: 25315430 DOI: 10.1158/1940-6207.capr-14-0132] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chromosome 8q24 has emerged as an important region for genetic susceptibility to various cancers, but little is known about the contribution of DNA methylation at 8q24. To evaluate variability in DNA methylation levels at 8q24 and the relationship with cancer susceptibility single nucleotide polymorphisms (SNPs) in this region, we quantified DNA methylation levels in peripheral blood at 145 CpG sites nearby 8q24 cancer susceptibility SNPs or MYC using pyrosequencing among 80 Caucasian men in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. For the 60 CpG sites meeting quality control, which also demonstrated temporal stability over a 5-year period, we calculated pairwise Spearman correlations for DNA methylation levels at each CpG site with 42 8q24 cancer susceptibility SNPs. To account for multiple testing, we adjusted P values into q values reflecting the false discovery rate (FDR). In contrast to the MYC CpG sites, most sites nearby the SNPs demonstrated good reproducibility, high methylation levels, and moderate-high between-individual variation. We observed 10 statistically significant (FDR < 0.05) CpG site-SNP correlations. These included correlations between an intergenic CpG site at Chr8:128393157 and the prostate cancer SNP rs16902094 (ρ = -0.54; P = 9.7 × 10(-7); q = 0.002), a PRNCR1 CpG site at Chr8:128167809 and the prostate cancer SNP rs1456315 (ρ = 0.52; P = 1.4 × 10(-6); q = 0.002), and two POU5F1B CpG sites and several prostate/colorectal cancer SNPs (for Chr8:128498051 and rs6983267, ρ = 0.46; P = 2.0 × 10(-5); q = 0.01). This is the first report of correlations between blood DNA methylation levels and cancer susceptibility SNPs at 8q24, suggesting that DNA methylation at this important susceptibility locus may contribute to cancer risk.
Collapse
Affiliation(s)
- Kathryn Hughes Barry
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland.
| | - Lee E Moore
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland
| | - Joshua Sampson
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland
| | - Liying Yan
- EpigenDx, Inc., Hopkinton, Massachusetts
| | - Ann Meyer
- EpigenDx, Inc., Hopkinton, Massachusetts
| | - Andrew J Oler
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Charles C Chung
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland
| | - Zhaoming Wang
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland
| | - Meredith Yeager
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland
| | - Laufey Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland
| | - Sonja I Berndt
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland
| |
Collapse
|
47
|
Kim C, Bassig BA, Seow WJ, Hu W, Purdue MP, Shu XO, Huang WY, Liu CS, Cheng WL, Lin TT, Xiang YB, Ji BT, Gao YT, Chow WH, Männistö S, Weinstein SJ, Albanes D, Zheng W, Hosgood HD, Lim U, Rothman N, Lan Q. Pooled analysis of mitochondrial DNA copy number and lung cancer risk in three prospective studies. Cancer Epidemiol Biomarkers Prev 2014; 23:2977-80. [PMID: 25293879 DOI: 10.1158/1055-9965.epi-14-1070] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND We previously reported that higher levels of mitochondrial DNA copy number (mtDNA CN) were associated with lung cancer risk among male heavy smokers (i.e., ≥20 cigarettes per day) in the Alpha-Tocopherol Beta-Carotene (ATBC) study. Here, we present two additional prospective investigations nested in the Prostate, Lung, Colorectal, and Ovarian (PLCO) cancer screening trial and the Shanghai Women's Health Study (SWHS), and pooled with previously published data from ATBC. MATERIALS All DNA were extracted from peripheral whole blood samples using the phenol-chloroform method, and mtDNA CN was assayed by fluorescence-based qPCR. Multivariate unconditional logistic regression models were used to estimate ORs and 95% confidence intervals for the association of mtDNA CN and lung cancer risk. RESULTS Overall, mtDNA CN was not associated with lung cancer risk in the PLCO, SWHS, or pooled populations (all P trends > 0.42, P heterogeneity = 0.0001), and mtDNA CN was inversely associated with lung cancer risk among male smokers in PLCO, the opposite direction observed in ATBC. In addition, the mtDNA CN association observed among male heavy smokers in ATBC was the opposite direction in PLCO. CONCLUSIONS mtDNA CN was not consistently associated with lung cancer risk across three prospective study populations from Europe, Asia, and the United States. IMPACT This pooled study suggests no consistent association between prediagnostic mtDNA CN levels and lung cancer risk across several populations. Cancer Epidemiol Biomarkers Prev; 23(12); 2977-80. ©2014 AACR.
Collapse
Affiliation(s)
- Christopher Kim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Bryan A Bassig
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Wei Jie Seow
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Wei Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mark P Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Xiao-Ou Shu
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Chin-San Liu
- Neurology and Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Wen-Ling Cheng
- Neurology and Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Ta-Tsung Lin
- Neurology and Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Yong-Bing Xiang
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bu-Tian Ji
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Wong-Ho Chow
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Satu Männistö
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Stephanie J Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Wei Zheng
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - H Dean Hosgood
- Albert Einstein College of Medicine, Yeshiva University, Bronx, New York
| | - Unhee Lim
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
48
|
Kim C, Bassig BA, Seow WJ, Hu W, Purdue MP, Huang WY, Liu CS, Cheng WL, Männistö S, Vermeulen R, Weinstein SJ, Lim U, Hosgood HD, Bonner MR, Caporaso NE, Albanes D, Lan Q, Rothman N. Mitochondrial DNA copy number and chronic lymphocytic leukemia/small lymphocytic lymphoma risk in two prospective studies. Cancer Epidemiol Biomarkers Prev 2014; 24:148-53. [PMID: 25293880 DOI: 10.1158/1055-9965.epi-14-0753] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Mitochondrial DNA copy number (mtDNA CN) may be modified by mitochondria in response to oxidative stress. Previously, mtDNA CN was associated with non-Hodgkin lymphoma (NHL) risk, particularly chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL). We conducted a replication study in the Prostate, Lung, Colorectal, and Ovarian (PLCO) study and pooled with published ATBC (Alpha-Tocopherol, Beta-Carotene) data. METHODS In PLCO, 292 NHL cases (95 CLL/SLL cases) and 301 controls were pooled with 142 NHL cases (47 CLL/SLL cases) and 142 controls from ATBC. Subjects answered a questionnaire and provided blood. DNA was extracted from prediagnostic peripheral white blood, and mtDNA CN assayed by quantitative polymerase chain reaction. Unconditional logistic regression estimated mtDNA CN and NHL risk by odds ratios (OR) and 95% confidence intervals (95% CI). RESULTS Greater mtDNA CN was associated with increased risk of CLL/SLL among males in PLCO (3rd vs. 1st tertile: OR, 2.21; 95% CI, 1.03-4.72; Ptrend: 0.049) and pooled (T3 vs. T1: OR, 3.12; 95% CI, 1.72-5.68; Ptrend: 0.0002). Association was stronger among male smokers (Ptrend: <0.0001) and essentially identical for cases diagnosed <6, >6-8, and >8 years from blood draw (pooled: Pinteraction: 0.65). mtDNA CN and risk of other NHL subtypes and multiple myeloma showed no association. CONCLUSIONS AND IMPACT Mitochondrial DNA CN was associated with risk of CLL/SLL in males/male smokers. The risk was observed among cases diagnosed as long as 8 years after blood draw. These results suggest that higher mtDNA CN may reflect a process involved in CLL/SLL development.
Collapse
Affiliation(s)
- Christopher Kim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Bryan A Bassig
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Wei Jie Seow
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Wei Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mark P Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Chin-San Liu
- Neurology and Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Wen-Ling Cheng
- Neurology and Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Satu Männistö
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Stephanie J Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Unhee Lim
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - H Dean Hosgood
- Albert Einstein College of Medicine, Yeshiva University, Bronx, New York
| | - Matthew R Bonner
- Department of Epidemiology and Environmental Health, State University of New York at Buffalo, Buffalo, New York
| | - Neil E Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
49
|
Cross AJ, Moore SC, Boca S, Huang WY, Xiong X, Stolzenberg-Solomon R, Sinha R, Sampson JN. A prospective study of serum metabolites and colorectal cancer risk. Cancer 2014; 120:3049-57. [PMID: 24894841 PMCID: PMC5819589 DOI: 10.1002/cncr.28799] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 04/03/2014] [Accepted: 04/24/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Colorectal cancer is highly prevalent, and the vast majority of cases are thought to be sporadic, although few risk factors have been identified. Using metabolomics technology, our aim was to identify biomarkers prospectively associated with colorectal cancer. METHODS This study included 254 incident colorectal cancers and 254 matched controls nested in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. Serum samples were collected at baseline, and the mean length of follow-up was 8 years. Serum metabolites were analyzed by ultra-high performance liquid-phase chromatography with tandem mass spectrometry, and gas chromatography coupled with mass spectrometry. Conditional logistic regression was used to calculate odds ratios (OR) and 95% confidence intervals (CI) for metabolites above the limit of detection and present in at least 80% of participants. RESULTS A total of 676 serum metabolites were measured; of these, 447 were of known identity and 278 of these were present in >80% of individuals. Overall, there was no association between serum metabolites and colorectal cancer; however, some suggestive associations were observed between individual metabolites and colorectal cancer but none reached statistical significance after Bonferroni correction for multiple comparisons. For example, leucyl-leucine was inversely associated (OR comparing the 90th to the 10th percentile = 0.50; 95% CI = 0.32-0.80; P = .003). In sex-stratified analyses, serum glycochenodeoxycholate was positively associated with colorectal cancer among women (OR(90th vs.10th percentile) = 5.34; 95% CI = 2.09-13.68; P = .0001). CONCLUSIONS No overall associations were observed between serum metabolites and colorectal cancer, but serum glycochenodeoxycholate, a bile acid metabolite, was positively associated with colorectal cancer among women.
Collapse
Affiliation(s)
- Amanda J. Cross
- Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, W2 1PG, United Kingdom
| | - Steven C. Moore
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Rockville, MD, United States
| | - Simina Boca
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Rockville, MD, United States
| | - Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Rockville, MD, United States
| | - Xiaoqin Xiong
- Information Management Services, Inc., 3901 Calverton Blvd, Suite 200, Calverton MD 20705, United States
| | - Rachael Stolzenberg-Solomon
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Rockville, MD, United States
| | - Rashmi Sinha
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Rockville, MD, United States
| | - Joshua N. Sampson
- Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Rockville, MD, United States
| |
Collapse
|
50
|
Jacobs EJ, Newton CC, Silverman DT, Nogueira LM, Albanes D, Männistö S, Pollak M, Stolzenberg-Solomon RZ. Serum transforming growth factor-β1 and risk of pancreatic cancer in three prospective cohort studies. Cancer Causes Control 2014; 25:1083-91. [PMID: 24913781 PMCID: PMC5920694 DOI: 10.1007/s10552-014-0409-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 05/30/2014] [Indexed: 12/20/2022]
Abstract
PURPOSE Clinically evident chronic pancreatitis is a strong risk factor for pancreatic cancer. A small Japanese cohort study previously reported that pre-diagnostic serum transforming growth factor-β1 (TGF-β1) concentration, a potential marker of subclinical pancreatic inflammation, was associated with higher risk of pancreatic cancer. We further explored this association in a larger prospective study. METHODS Serum TGF-β1 concentrations were measured in pre-diagnostic samples from 729 pancreatic cancer cases and 907 matched controls from a cohort of Finnish male smokers (the Alpa-Tocopherol, Beta-Carotene (ATBC) Cancer Prevention Study) and two cohorts of US men and women, the Cancer Prevention Study-II and the Prostate Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial. Multivariable-adjusted odds ratios (ORs) were estimated using conditional logistic regression. RESULTS Overall, serum TGF-β1 concentration was not associated with a clear increase in pancreatic cancer risk (OR 1.36, 95 % confidence interval (CI) 0.98-1.88 for highest vs. lowest quintile, p trend = 0.20). However, this association differed significantly by follow-up time (p = 0.02). Serum TGF-β1 concentration was not associated with risk during the first 10 years of follow-up, but was associated with higher risk during follow-up after 10 years (OR 2.13, 95 % CI 1.23-3.68 for highest vs. lowest quintile, p trend = 0.001). During follow-up after 10 years, serum TGF-β1 was associated with higher risk only in the ATBC cohort, although most subjects were from ATBC during this time period and statistical evidence for heterogeneity across cohorts was limited (p = 0.14). CONCLUSIONS These results suggest that high serum TGF-β1 may be associated with increased risk of pancreatic cancer although a long follow-up period may be needed to observe this association.
Collapse
Affiliation(s)
- Eric J Jacobs
- Epidemiology Research Program, American Cancer Society, National Home Office, 250 Williams Street, Atlanta, GA, 30303-1002, USA,
| | | | | | | | | | | | | | | |
Collapse
|