1
|
Chlebowski RT, Aragaki AK, Pan K, Haque R, Rohan TE, Song M, Wactawski-Wende J, Lane DS, Harris HR, Strickler H, Kauntiz AM, Runowicz CD. Menopausal Hormone Therapy and Ovarian and Endometrial Cancers: Long-Term Follow-Up of the Women's Health Initiative Randomized Trials. J Clin Oncol 2024; 42:3537-3549. [PMID: 39173088 DOI: 10.1200/jco.23.01918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 04/18/2024] [Accepted: 05/21/2024] [Indexed: 08/24/2024] Open
Abstract
PURPOSE Menopausal hormone therapy's influence on ovarian and endometrial cancers remains unsettled. Therefore, we assessed the long-term influence of conjugated equine estrogen (CEE) plus medroxyprogesterone acetate (MPA) and CEE-alone on ovarian and endometrial cancer incidence and mortality in the Women's Health Initiative randomized, placebo-controlled clinical trials. MATERIALS AND METHODS Postmenopausal women, age 50-79 years, were entered on two randomized clinical trials evaluating different menopausal hormone therapy regimens. In 16,608 women with a uterus, 8,506 were randomly assigned to once daily 0.625 mg of CEE plus 2.5 mg once daily of MPA and 8,102 placebo. In 10,739 women with previous hysterectomy, 5,310 were randomly assigned to once daily 0.625 mg of CEE-alone and 5,429 placebo. Intervention was stopped for cause before planned 8.5-year intervention after 5.6 years (CEE plus MPA) and after 7.2 years (CEE-alone). Outcomes include incidence and mortality from ovarian and endometrial cancers and deaths after these cancers. RESULTS After 20-year follow-up, CEE-alone, versus placebo, significantly increased ovarian cancer incidence (35 cases [0.041%] v 17 [0.020%]; hazard ratio [HR], 2.04 [95% CI, 1.14 to 3.65]; P = .014) and ovarian cancer mortality (P = .006). By contrast, CEE plus MPA, versus placebo, did not increase ovarian cancer incidence (75 cases [0.051%] v 63 [0.045%]; HR, 1.14 [95% CI, 0.82 to 1.59]; P = .44) or ovarian cancer mortality but did significantly lower endometrial cancer incidence (106 cases [0.073%] v 140 [0.10%]; HR, 0.72 [95% CI, 0.56 to 0.92]; P = .01). CONCLUSION In randomized clinical trials, CEE-alone increased ovarian cancer incidence and ovarian cancer mortality, while CEE plus MPA did not. By contrast, CEE plus MPA significantly reduced endometrial cancer incidence.
Collapse
Affiliation(s)
| | - Aaron K Aragaki
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA
| | - Kathy Pan
- Kaiser Permanente Southern California, Downey, CA
| | - Reina Haque
- Department of Health Systems Science, Kaiser Permanente Bernard J Tyson School of Medicine, Pasadena, CA
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Mihae Song
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, University at Buffalo, Buffalo, NY
| | - Dorothy S Lane
- Department of Epidemiology and Environmental Health, University at Buffalo, Buffalo, NY
- Department of Family, Population and Preventive Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY
| | - Holly R Harris
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA
| | - Howard Strickler
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Andrew M Kauntiz
- Department of Obstetrics & Gynecology, University of Florida College of Medicine-Jacksonville, Jacksonville, FL
| | - Carolyn D Runowicz
- Florida International University Herbert Wertheim College of Medicine, Miami, FL
| |
Collapse
|
2
|
Denos M, Sun YQ, Brumpton BM, Li Y, Albanes D, Burnett-Hartman A, Campbell PT, Küry S, Li CI, White E, Samadder JN, Jenkins MA, Mai XM. Sex hormones and risk of lung and colorectal cancers in women: a Mendelian randomization study. Sci Rep 2024; 14:23891. [PMID: 39396092 PMCID: PMC11470916 DOI: 10.1038/s41598-024-75305-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024] Open
Abstract
The roles of sex hormones such as estradiol, testosterone, and sex hormone-binding globulin (SHBG) in the etiology of lung and colorectal cancers in women, among the most common cancers after breast cancer, are unclear. This Mendelian randomization (MR) study evaluated such potential causal associations in women of European ancestry. We used summary statistics data from genome-wide association studies on sex hormones and from the Trøndelag Health Study (HUNT) and large consortia on cancers. There was suggestive evidence of 1-standard deviation increase in total testosterone levels being associated with a lower risk of lung non-adenocarcinoma (hazard ratio 0.60, 95% confidence interval 0.37-0.98) in the HUNT Study. However, this was not confirmed by using data from a larger consortium. In general, we did not find convincing evidence to support a causal role of sex hormones on risk of lung and colorectal cancers in women of European ancestry.
Collapse
Affiliation(s)
- Marion Denos
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Yi-Qian Sun
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Pathology, Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim, Norway
- Center for Oral Health Services and Research Mid-Norway (TkMidt), Trondheim, Norway
| | - Ben Michael Brumpton
- Department of Public Health and Nursing, K.G. Jebsen Centre for Genetic Epidemiology, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Public Health and Nursing, HUNT Research Centre, NTNU, Norwegian University of Science and Technology, Levanger, 7600, Norway
- Clinic of Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Yafang Li
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Peter T Campbell
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sébastien Küry
- Service de Génétique Médicale, Nantes Université, CHU Nantes, Nantes, 44000, France
| | - Christopher I Li
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Emily White
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jewel N Samadder
- Department of Gastroenterology and Hepatology, Mayo Clinic, Scottsdale, AZ, USA
| | - Mark A Jenkins
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Xiao-Mei Mai
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
3
|
Huang YX, Wu JH, Zhao YQ, Sui WN, Tian T, Han WX, Ni J. An atlas on risk factors for gastrointestinal cancers: A systematic review of Mendelian randomization studies. Prev Med 2024; 189:108147. [PMID: 39368643 DOI: 10.1016/j.ypmed.2024.108147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/07/2024]
Abstract
OBJECTIVE Gastrointestinal cancers are one of the most frequent cancer types and seriously threaten human life and health. Recent studies attribute the occurrence of gastrointestinal cancers to both genetic and environmental factors, yet the intrinsic etiology remains unclear. Mendelian randomization is a powerful well-established statistical method that is based on genome-wide association study (GWAS) to evaluate the causal relationship between exposures and outcomes. In the present study, we aimed to conduct a systematic review of Mendelian randomization studies investigating any causal risk factors for gastrointestinal cancers. METHODS We systematically searched Mendelian randomization studies that addressed the associations of genetically predicted exposures with five main gastrointestinal cancers from September 2014 to March 2024, as well as testing the research quality and validity. RESULTS Our findings suggested robust and consistent causal effects of body mass index (BMI), basal metabolic rate, fatty acids, total cholesterol, total bilirubin, insulin like growth factor-1, eosinophil counts, interleukin 2, alcohol consumption, coffee consumption, apolipoprotein B on colorectal cancer risks, BMI, waist circumference, low-density lipoprotein (LDL), total testosterone, smoking on gastric cancer risks, BMI, fasting insulin, LDL, waist circumference, visceral adipose tissue (VAT), immune cells, type 2 diabetes mellitus (T2DM) on pancreatic cancer risks, waist circumference, smoking, T2DM on esophageal adenocarcinoma risks, and VAT, ferritin, transferrin, alcohol consumption, hepatitis B virus infection, rheumatoid arthritis on liver cancer risks, respectively. CONCLUSION Larger, well-designed Mendelian randomization studies are practical in determining the causal status of risk factors for diseases.
Collapse
Affiliation(s)
- Yi-Xuan Huang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Jun-Hua Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Yu-Qiang Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Wan-Nian Sui
- Department of Gastrointestinal Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tian Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Wen-Xiu Han
- Department of Gastrointestinal Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Jing Ni
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.
| |
Collapse
|
4
|
Liu Z, Jiang M, Wang T, Li F, Zhu Y. A cause-effect relationship between uterine diseases and breast cancer: A bidirectional Mendelian randomization study. Heliyon 2024; 10:e38130. [PMID: 39364229 PMCID: PMC11447327 DOI: 10.1016/j.heliyon.2024.e38130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024] Open
Abstract
Objective To explore the cause-effect relationship between uterine diseases (UDs) and breast cancer (BC) and underlying mechanism of the cause-effect relationship, enhance understanding of the association between BC and UDs. Methods A two-sample bidirectional Mendelian randomization (MR) analysis was conducted. We obtained summary statistics data from GWAS for BC, endometriosis, endometrial cancer (EC), uterine leiomyoma (UL), uterine polyps (UP), and cervical cancer (CC). Independent SNPs were selected as instrumental variables (IVs) for each disease. The inverse variance weighted (IVW) method was primary used for estimating the causal association between UDs and BC. To further evaluate the consistency and dependability of the results, we also utilized the weighted median, weighted mode, simple mode, and MR-Egger methods, along with sensitivity analyses. Furthermore, a supplementary analysis focusing on the variants linked to BC and UDs was conducted. This involved identifying corresponding genes and subsequently performing KEGG/GO analyses to investigate potential molecular mechanisms. Results The results indicated significant associations between genetic susceptibility to endometriosis, EC, and UL with BC risk. The odds ratios (ORs) were as follows: endometriosis at 0.963 (95 % CI, 0.942-0.984; p = 7.11e-5), EC at 1.056 (95 % CI, 1.033-1.081; p = 2.39e-6), and UL at 1.027 (95 % CI, 1.006-1.048; p = 0.010). Conversely, the predisposition to BC inferred from genetic factors was markedly correlated with an elevated risk of EC indicated by an OR of 1.066 (95 % CI, 1.019-1.116; p = 0.006), and was correlated with UP risk (OR, 1.001,95 % CI, 1.000-1.002; p = 0.001).Sensitivity analyses provided weak evidence for these effects, suggesting that the study's outcomes are consistent and trustworthy. Further analysis of the genetic variants associated with BC, and these related genes are enriched in Cellular senescence, GnRH secretion, Phosphatidylinositol signaling system, and so on. Conclusion This study corroborates the existence of a reciprocal causal relationship between BC and EC, as well as highlighting the substantial correlations between a genetic susceptibility to UL and endometriosis with BC. BC may exert their influence on EC and UP through Cellular senescence, GnRH secretion, and other pathways. These discoveries offer fresh perspectives on the genetic pathogenesis of BC and UDs, and can guide future experimental studies. Additionally, they lay down a groundwork for the development of tailored preventative and therapeutic strategies moving forward.
Collapse
Affiliation(s)
- Zhipeng Liu
- Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou, Jiangsu, 225300, China
| | - Min Jiang
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, 225300, China
| | - Taiyu Wang
- Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou, Jiangsu, 225300, China
| | - Fang Li
- Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou, Jiangsu, 225300, China
| | - Yinxing Zhu
- Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou, Jiangsu, 225300, China
| |
Collapse
|
5
|
Ji L, Liu Y, Wang Z, Huang Q, Cai J, Gu H, Li J, Chen X, Feng C, He X, Deng X, Cheng X, Kong X, Zhu X, Wu T, Yang B, Lin Z, Yang X, Feng G, Yu J. Causal effect analysis of estrogen receptor associated breast cancer and clear cell ovarian cancer. Am J Transl Res 2024; 16:2699-2710. [PMID: 39006281 PMCID: PMC11236669 DOI: 10.62347/ecoo9552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Evidence indicates that the risk of developing a secondary ovarian cancer (OC) is correlated with estrogen receptor (ER) status. However, the clinical significance of the relationship between ER-associated breast cancer (BC) and clear cell ovarian cancer (CCOC) remains elusive. METHODS Independent single nucleotide polymorphisms (SNPs) strongly correlated with exposure were extracted, and those associated with confounders and outcomes were removed using the PhenoScanner database. SNP effects were extracted from the outcome datasets with minor allele frequency > 0.01 as the filtration criterion. Next, valid instrumental variables (IVs) were obtained by harmonizing exposure and outcome effects and further filtered based on F-statistics (> 10). Mendelian randomization (MR) assessment of valid IVs was carried out using inverse variance weighted (IVW), MR Egger (ME), weighted median (WM), and multiplicative random effects-inverse variance weighted (MRE-IVW) methods. For sensitivity analysis and visualization of MR findings, a heterogeneity test, a pleiotropy test, a leave-one-out test, scatter plots, forest plots, and funnel plots were employed. RESULTS MR analyses with all four methods revealed that CCOC was not causally associated with ER-negative BC (IVW results: odds ratio (OR) = 0.89, 95% confidence interval (CI) = 0.66-1.20, P = 0.431) or ER-positive BC (IVW results: OR = 0.99, 95% CI = 0.88-1.12, P = 0.901). F-statistics were computed for each valid IV, all of which exceeded 10. The stability and reliability of the results were confirmed by sensitivity analysis. CONCLUSIONS Our findings indicated that CCOC dids not have a causal association with ER-associated BC. The absence of a definitive causal link between ER-associated BC and CCOC suggested a minimal true causal influence of ER-associated BC exposure factors on CCOC. These results indicated that individuals afflicted by ER-associated BC could alleviate concerns regarding the developing of CCOC, thereby aiding in preserving their mental well-being stability and optimizing the efficacy of primary disease treatment.
Collapse
Affiliation(s)
- Li Ji
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Yanbo Liu
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical UniversitySuzhou 215000, Jiangsu, China
| | - Zihan Wang
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Qiuru Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Jiaying Cai
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Han Gu
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Jiaxin Li
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Xia Chen
- Department of Obstetrics and Gynecology, Nantong First People’s Hospital, Affiliated Hospital 2 of Nantong University, Nantong UniversityNantong 226001, Jiangsu, China
| | - Chenrui Feng
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Xuxin He
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Xiaonan Deng
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Xinmeng Cheng
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Xiuwen Kong
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Xiaoqi Zhu
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Tong Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Binbin Yang
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Ziwen Lin
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| | - Xiaoqing Yang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Nantong University, Nantong UniversityNantong 226001, Jiangsu, China
| | - Guannan Feng
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical UniversitySuzhou 215000, Jiangsu, China
| | - Jun Yu
- Institute of Reproductive Medicine, School of Medicine, Nantong UniversityNantong 226001, Jiangsu, China
| |
Collapse
|
6
|
Caerts D, Garmyn M, Güvenç C. A Narrative Review of the Role of Estrogen (Receptors) in Melanoma. Int J Mol Sci 2024; 25:6251. [PMID: 38892441 PMCID: PMC11173079 DOI: 10.3390/ijms25116251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
In this narrative review, we attempt to provide an overview of the evidence regarding the role of estrogen (receptors) in cutaneous melanoma (CM). We reviewed 68 studies and 4 systematic reviews and meta-analyses published from 2002 up to and including 2022. The prevailing presence of estrogen receptor β (ERβ) instead of estrogen receptor α (ERα) in CM is notable, with ERβ potentially playing a protective role and being less frequently detected in progressive cases. While men with CM generally experience a less favorable prognosis, this distinction may become negligible with advancing age. The role of oral contraceptives (OC) and hormone replacement therapy (HRT) in CM remains controversial. However, recent studies tend to associate the use of these exogenous hormones with a heightened risk of CM, mostly only when using estrogen therapy and not in combination with progesterone. On the contrary, the majority of studies find no substantial influence of in vitro fertilization (IVF) treatment on CM risk. Reproductive factors, including younger age at first childbirth, higher parity, and shorter reproductive life, show conflicting evidence, with some studies suggesting a lower CM risk. We suggest an important role for estrogens in CM. More research is needed, but the integration of estrogens and targeting the estrogen receptors in melanoma therapy holds promise for future developments in the field.
Collapse
Affiliation(s)
| | | | - Canan Güvenç
- Department of Dermatology, University Hospitals Leuven, 3000 Leuven, Belgium; (D.C.); (M.G.)
| |
Collapse
|
7
|
Liang H, Geng S, Wang Y, Fang Q, Xin Y, Li Y. Tumour-derived exosome SNHG17 induced by oestrogen contributes to ovarian cancer progression via the CCL13-CCR2-M2 macrophage axis. J Cell Mol Med 2024; 28:e18315. [PMID: 38680032 PMCID: PMC11056704 DOI: 10.1111/jcmm.18315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 05/01/2024] Open
Abstract
Oestrogen is known to be strongly associated with ovarian cancer. There was much work to show the importance of lncRNA SNHG17 in ovarian cancer. However, no study has revealed the molecular regulatory mechanism and functional effects between oestrogen and SNHG17 in the development and metastasis of ovarian cancer. In this study, we found that SNHG17 expression was significantly increased in ovarian cancer and positively correlated with oestrogen treatment. Oestrogen could promote M2 macrophage polarization as well as ovarian cancer cells SKOV3 and ES2 cell exosomal SNHG17 expression. When exposure to oestrogen, exosomal SNHG17 promoted ovarian cancer cell proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) in vitro, and tumour growth and lung metastasis in vivo by accelerating M2-like phenotype of macrophages. Mechanically, exosomal SNHG17 could facilitate the release of CCL13 from M2 macrophage via the PI3K-Akt signalling pathway. Moreover, CCL13-CCR2 axis was identified to be involved in ovarian cancer tumour behaviours driven by oestrogen. There results demonstrate a novel mechanism that exosomal SNHG17 exerts an oncogenic effect on ovarian cancer via the CCL13-CCR2-M2 macrophage axis upon oestrogen treatment, of which SNHG17 may be a potential biomarker and therapeutic target for ovarian cancer responded to oestrogen.
Collapse
Affiliation(s)
- Haiyan Liang
- Department of Obstetrics and GynecologyChina‐Japan Friendship HospitalBeijingChina
| | - Shuo Geng
- Department of Obstetrics and GynecologyChina‐Japan Friendship HospitalBeijingChina
| | - Yadong Wang
- Scientific Research DepartmentGeneX Health Co., LtdBeijingChina
| | - Qing Fang
- Institute of Clinical MedicineChina‐Japan Friendship HospitalBeijingChina
| | - Yongfeng Xin
- Department of GynecologyThe People's Hospital of DaLaTeOrdosInner MongoliaChina
| | - Yanqing Li
- Department of GynecologyHebei Provincial Hospital of Traditional Chinese MedicineWuhanHebeiChina
| |
Collapse
|
8
|
Denos M, Sun YQ, Brumpton B, Li Y, Albanes D, Burnett-Hartman A, Campbell PT, Küry S, Li CI, White E, Samadder JN, Jenkins M, Mai XM. Sex hormones and risk of lung and colorectal cancers in women: a Mendelian randomization study. RESEARCH SQUARE 2024:rs.3.rs-4083598. [PMID: 38659935 PMCID: PMC11042402 DOI: 10.21203/rs.3.rs-4083598/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The roles of sex hormones such as estradiol, testosterone, and sex hormone-binding globulin (SHBG) in the etiology of lung and colorectal cancers in women, among the most common cancers after breast cancer, are unclear. This Mendelian randomization (MR) study evaluated such potential causal associations in women of European ancestry. We used summary statistics data from genome-wide association studies (GWASs) on sex hormones and from the Trøndelag Health (HUNT) Study and large consortia on cancers. There was suggestive evidence of genetically predicted 1-standard deviation increase in total testosterone levels being associated with a lower risk of lung non-adenocarcinoma (hazard ratio (HR) 0.60, 95% CI 0.37-0.98) in the HUNT Study. However, this was not confirmed by using data from a larger consortium. In general, we did not find convincing evidence to support a causal role of sex hormones on risk of lung and colorectal cancers in women of European ancestry.
Collapse
Affiliation(s)
| | - Yi-Qian Sun
- Norwegian University of Science and Technology
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Watts EL, Moore SC, Gunter MJ, Chatterjee N. Adiposity and cancer: meta-analysis, mechanisms, and future perspectives. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.16.24302944. [PMID: 38405761 PMCID: PMC10889047 DOI: 10.1101/2024.02.16.24302944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Obesity is a recognised risk factor for many cancers and with rising global prevalence, has become a leading cause of cancer. Here we summarise the current evidence from both population-based epidemiologic investigations and experimental studies on the role of obesity in cancer development. This review presents a new meta-analysis using data from 40 million individuals and reports positive associations with 19 cancer types. Utilising major new data from East Asia, the meta-analysis also shows that the strength of obesity and cancer associations varies regionally, with stronger relative risks for several cancers in East Asia. This review also presents current evidence on the mechanisms linking obesity and cancer and identifies promising future research directions. These include the use of new imaging data to circumvent the methodological issues involved with body mass index and the use of omics technologies to resolve biologic mechanisms with greater precision and clarity.
Collapse
Affiliation(s)
- Eleanor L Watts
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Shady Grove, MD, USA
| | - Steven C Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Shady Grove, MD, USA
| | - Marc J Gunter
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Nilanjan Chatterjee
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, USA
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, USA
| |
Collapse
|
10
|
Costas L, Frias-Gomez J, Peinado FM, Molina-Molina JM, Peremiquel-Trillas P, Paytubi S, Crous-Bou M, de Francisco J, Caño V, Benavente Y, Pelegrina B, Martínez JM, Pineda M, Brunet J, Matias-Guiu X, de Sanjosé S, Ponce J, Olea N, Alemany L, Fernández MF. Total Effective Xenoestrogen Burden in Serum Samples and Risk of Endometrial Cancer in the Spanish Screenwide Case-Control Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:27012. [PMID: 38415615 PMCID: PMC10901108 DOI: 10.1289/ehp13202] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 01/08/2024] [Accepted: 01/23/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Endometrial cancer is a hormone-dependent cancer, and estrogens play a relevant role in its etiology. However, little is known about the effects of environmental pollutants that act as xenoestrogens or that influence estrogenic activity through different pathways. OBJECTIVE We aimed to assess the relationship between the combined estrogenic activity of mixtures of xenoestrogens present in serum samples and the risk of endometrial cancer in the Screenwide case-control study. METHODS The total effective xenoestrogen burden (TEXB) attributable to organohalogenated compounds (TEXB- α ) and to endogenous hormones and more polar xenoestrogens (TEXB- β ) was assessed in serum from 156 patients with endometrial cancer (cases) and 150 controls by combining chemical extraction and separation by high-performance liquid chromatography with the E-SCREEN bioassay for estrogenicity. RESULTS Median TEXB- α and TEXB- β levels for cases (0.30 and 1.25 Eeq pM / mL , respectively) and controls (0.42 and 1.28 Eeq pM / mL , respectively) did not significantly differ (p = 0.653 and 0.933, respectively). An inverted-U risk trend across serum TEXB- α and TEXB- β levels was observed in multivariate adjusted models: Positive associations were observed for the second category of exposure in comparison to the lowest category of exposure [odds ratio ( OR ) = 2.11 (95% CI: 1.13, 3.94) for TEXB- α , and OR = 3.32 (95% CI: 1.62, 6.81) for TEXB- β ], whereas no significant associations were observed between the third category of exposure and the first [OR = 1.22 (95% CI: 0.64, 2.31) for TEXB- α , and OR = 1.58 (95% CI: 0.75, 3.33) for TEXB- β ]. In mutually adjusted models for TEXB- α and TEXB- β levels, the association of TEXB- α with endometrial cancer risk was attenuated [OR = 1.45 (95% CI: 0.61, 3.47) for the second category of exposure], as well as estimates for TEXB- β (OR = 2.68 ; 95% CI: 1.03, 6.99). Most of the individual halogenated contaminants showed no associations with both TEXB and endometrial cancer. CONCLUSIONS We evaluated serum total xenoestrogen burden in relation to endometrial cancer risk and found an inverted-U risk trend across increasing categories of exposure. The use of in vitro bioassays with human samples may lead to a paradigm shift in the way we understand the negative impact of chemical mixtures on human health effects. These results are relevant from a public health perspective and for decision-makers in charge of controlling the production and distribution of chemicals with xenoestrogenic activity. https://doi.org/10.1289/EHP13202.
Collapse
Affiliation(s)
- Laura Costas
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, Hospitalet de Llobregat, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Jon Frias-Gomez
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, Hospitalet de Llobregat, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- University of Barcelona, Barcelona, Spain
| | - Francisco M. Peinado
- Centre of Biomedical Research, University of Granada, Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), Hospital Universitario San Cecilio, Granada, Spain
| | | | - Paula Peremiquel-Trillas
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, Hospitalet de Llobregat, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- University of Barcelona, Barcelona, Spain
| | - Sonia Paytubi
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, Hospitalet de Llobregat, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Marta Crous-Bou
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, Hospitalet de Llobregat, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Javier de Francisco
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Department of Anesthesiology, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Victor Caño
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Department of Anesthesiology, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Yolanda Benavente
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, Hospitalet de Llobregat, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Beatriz Pelegrina
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, Hospitalet de Llobregat, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - José Manuel Martínez
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Department of Gynecology, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Marta Pineda
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Hereditary Cancer group, Molecular Mechanisms and Experimental Therapy in Oncology Program, Catalan Institute of Oncology, Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Cancer (CIBERONC), Carlos III Institute of Health, Madrid, Spain
| | - Joan Brunet
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Hereditary Cancer group, Molecular Mechanisms and Experimental Therapy in Oncology Program, Catalan Institute of Oncology, Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Cancer (CIBERONC), Carlos III Institute of Health, Madrid, Spain
| | - Xavier Matias-Guiu
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Cancer (CIBERONC), Carlos III Institute of Health, Madrid, Spain
- Department of Pathology, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Silvia de Sanjosé
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Barcelona Institute for Global Health, Barcelona, Spain
| | - Jordi Ponce
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Department of Gynecology, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Nicolás Olea
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Centre of Biomedical Research, University of Granada, Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), Hospital Universitario San Cecilio, Granada, Spain
| | - Laia Alemany
- Cancer Epidemiology Research Programme, Catalan Institute of Oncology, Hospitalet de Llobregat, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Mariana F. Fernández
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Centre of Biomedical Research, University of Granada, Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), Hospital Universitario San Cecilio, Granada, Spain
| |
Collapse
|
11
|
Burgess S, Cronjé HT. Incorporating biological and clinical insights into variant choice for Mendelian randomisation: examples and principles. EGASTROENTEROLOGY 2024; 2:e100042. [PMID: 38362310 PMCID: PMC7615644 DOI: 10.1136/egastro-2023-100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Mendelian randomisation is an accessible and valuable epidemiological approach to provide insight into the causal nature of relationships between risk factor exposures and disease outcomes. However, if performed without critical thought, we may simply have replaced one set of implausible assumptions (no unmeasured confounding or reverse causation) with another set of implausible assumptions (no pleiotropy or other instrument invalidity). The most critical decision to avoid pleiotropy is which genetic variants to use as instrumental variables. Two broad strategies for instrument selection are a biologically motivated strategy and a genome-wide strategy; in general, a biologically motivated strategy is preferred. In this review, we discuss various ways of implementing a biologically motivated selection strategy: using variants in a coding gene region for the exposure or a gene region that encodes a regulator of exposure levels, using a positive control variable and using a biomarker as the exposure rather than its behavioural proxy. In some cases, a genome-wide analysis can provide important complementary evidence, even when its reliability is questionable. In other cases, a biologically-motivated analysis may not be possible. The choice of genetic variants must be informed by biological and functional considerations where possible, requiring collaboration to combine biological and clinical insights with appropriate statistical methodology.
Collapse
Affiliation(s)
- Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Héléne Toinét Cronjé
- Health Analytics, Lane Clark & Peacock LLP, London, UK
- Department of Public Health, Section of Epidemiology, University of Copenhagen, København, Denmark
| |
Collapse
|
12
|
Larsson SC, Spyrou N, Mantzoros CS. Body fatness associations with cancer: evidence from recent epidemiological studies and future directions. Metabolism 2022; 137:155326. [PMID: 36191637 DOI: 10.1016/j.metabol.2022.155326] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 12/12/2022]
Abstract
This narrative review highlights current evidence linking greater body fatness to risk of various cancers, with focus on evidence from recent large cohort studies and pooled analyses of cohort studies as well as Mendelian randomization studies (which utilized genetic variants associated with body mass index to debrief the causal effect of higher body fatness on cancer risk). This review also provides insights into the biological mechanisms underpinning the associations. Data from both observational and Mendelian randomization studies support the associations of higher body mass index with increased risk of many cancers with the strongest evidence for digestive system cancers, including esophageal, stomach, colorectal, liver, gallbladder, and pancreatic cancer, as well as kidney, endometrial, and ovarian (weak association) cancer. Evidence from observational studies suggests that greater body fatness has contrasting effects on breast cancer risk depending on menopausal status and on prostate cancer risk depending on disease stage. Experimental and Mendelian randomization studies indicate that adiponectin, insulin, and sex hormone pathways play an important role in mediating the link between body fatness and cancer risk. The possible role of specific factors and pathways, such as other adipocytokines and hormones and the gut microbiome in mediating the associations between greater body fatness and cancer risk is yet uncertain and needs investigation in future studies. With rising prevalence of overweight and obesity worldwide, the proportion of cancer caused by excess body fatness is expected to increase. There is thus an urgent need to identify efficient ways at the individual and societal level to improve diet and physical activity patterns to reduce the burden of obesity and accompanying comorbidities, including cancer.
Collapse
Affiliation(s)
- Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| | - Nikolaos Spyrou
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA; Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Cheminformatics analysis of chemicals that increase estrogen and progesterone synthesis for a breast cancer hazard assessment. Sci Rep 2022; 12:20647. [PMID: 36450809 PMCID: PMC9712655 DOI: 10.1038/s41598-022-24889-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Factors that increase estrogen or progesterone (P4) action are well-established as increasing breast cancer risk, and many first-line treatments to prevent breast cancer recurrence work by blocking estrogen synthesis or action. In previous work, using data from an in vitro steroidogenesis assay developed for the U.S. Environmental Protection Agency (EPA) ToxCast program, we identified 182 chemicals that increased estradiol (E2up) and 185 that increased progesterone (P4up) in human H295R adrenocortical carcinoma cells, an OECD validated assay for steroidogenesis. Chemicals known to induce mammary effects in vivo were very likely to increase E2 or P4 synthesis, further supporting the importance of these pathways for breast cancer. To identify additional chemical exposures that may increase breast cancer risk through E2 or P4 steroidogenesis, we developed a cheminformatics approach to identify structural features associated with these activities and to predict other E2 or P4 steroidogens from their chemical structures. First, we used molecular descriptors and physicochemical properties to cluster the 2,012 chemicals screened in the steroidogenesis assay using a self-organizing map (SOM). Structural features such as triazine, phenol, or more broadly benzene ramified with halide, amine or alcohol, are enriched for E2 or P4up chemicals. Among E2up chemicals, phenol and benzenone are found as significant substructures, along with nitrogen-containing biphenyls. For P4up chemicals, phenol and complex aromatic systems ramified with oxygen-based groups such as flavone or phenolphthalein are significant substructures. Chemicals that are active for both E2up and P4up are enriched with substructures such as dihydroxy phosphanedithione or are small chemicals that contain one benzene ramified with chlorine, alcohol, methyl or primary amine. These results are confirmed with a chemotype ToxPrint analysis. Then, we used machine learning and artificial intelligence algorithms to develop and validate predictive classification QSAR models for E2up and P4up chemicals. These models gave reasonable external prediction performances (balanced accuracy ~ 0.8 and Matthews Coefficient Correlation ~ 0.5) on an external validation. The QSAR models were enriched by adding a confidence score that considers the chemical applicability domain and a ToxPrint assessment of the chemical. This profiling and these models may be useful to direct future testing and risk assessments for chemicals related to breast cancer and other hormonally-mediated outcomes.
Collapse
|
14
|
D'Urso S, Arumugam P, Weider T, Hwang LD, Bond TA, Kemp JP, Warrington NM, Evans DM, O'Mara TA, Moen GH. Mendelian randomization analysis of factors related to ovulation and reproductive function and endometrial cancer risk. BMC Med 2022; 20:419. [PMID: 36320039 PMCID: PMC9623961 DOI: 10.1186/s12916-022-02585-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Observational epidemiological studies suggest a link between several factors related to ovulation and reproductive function and endometrial cancer (EC) risk; however, it is not clear whether these relationships are causal, and whether the risk factors act independently of each other. The aim of this study was to investigate putative causal relationships between the number of live births, age at last live birth, and years ovulating and EC risk. METHODS: We conducted a series of observational analyses to investigate various risk factors and EC risk in the UK Biobank (UKBB). Additionally, multivariate analysis was performed to elucidate the relationship between the number of live births, age at last live birth, and years ovulating and other related factors such as age at natural menopause, age at menarche, and body mass index (BMI). Secondly, we used Mendelian randomization (MR) to assess if these observed relationships were causal. Genome-wide significant single nucleotide polymorphisms (SNPs) were extracted from previous studies of woman's number of live births, age at menopause and menarche, and BMI. We conducted a genome-wide association analysis using the UKBB to identify SNPs associated with years ovulating, years using the contraceptive pill, and age at last live birth. RESULTS We found evidence for a causal effect of the number of live births (inverse variance weighted (IVW) odds ratio (OR): 0.537, p = 0.006), the number of years ovulating (IVW OR: 1.051, p = 0.014), in addition to the known risk factors BMI, age at menarche, and age at menopause on EC risk in the univariate MR analyses. Due to the close relationships between these factors, we followed up with multivariable MR (MVMR) analysis. Results from the MVMR analysis showed that number of live births had a causal effect on EC risk (OR: 0.783, p = 0.036) independent of BMI, age at menarche and age at menopause. CONCLUSIONS MVMR analysis showed that the number of live births causally reduced the risk of EC.
Collapse
Affiliation(s)
- Shannon D'Urso
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Pooja Arumugam
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Therese Weider
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Liang-Dar Hwang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Tom A Bond
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
- Bristol Medical School, Population Health Science, University of Bristol, Bristol, UK
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - John P Kemp
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Nicole M Warrington
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - David M Evans
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Tracy A O'Mara
- Cancer Research Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Gunn-Helen Moen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia.
- Bristol Medical School, Population Health Science, University of Bristol, Bristol, UK.
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.
| |
Collapse
|
15
|
Peduzzi G, Archibugi L, Katzke V, Gentiluomo M, Capurso G, Milanetto AC, Gazouli M, Goetz M, Brenner H, Vermeulen RCH, Talar-Wojnarowska R, Vanella G, Tavano F, Lucchesi M, Mohelnikova-Duchonova B, Chen X, Kiudelis V, Hegyi P, Oliverius M, Stocker H, Stornello C, Vodickova L, Souček P, Neoptolemos JP, Testoni SGG, Morelli L, Lawlor RT, Basso D, Izbicki JR, Ermini S, Kupcinskas J, Pezzilli R, Boggi U, van Laarhoven HWM, Szentesi A, Erőss B, Capretti G, Schöttker B, Skieceviciene J, Aoki MN, van Eijck CHJ, Cavestro GM, Canzian F, Campa D. Common variability in oestrogen-related genes and pancreatic ductal adenocarcinoma risk in women. Sci Rep 2022; 12:18100. [PMID: 36302831 PMCID: PMC9613634 DOI: 10.1038/s41598-022-22973-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 10/21/2022] [Indexed: 12/30/2022] Open
Abstract
The incidence of pancreatic ductal adenocarcinoma (PDAC) is different among males and females. This disparity cannot be fully explained by the difference in terms of exposure to known risk factors; therefore, the lower incidence in women could be attributed to sex-specific hormones. A two-phase association study was conducted in 12,387 female subjects (5436 PDAC cases and 6951 controls) to assess the effect on risk of developing PDAC of single nucleotide polymorphisms (SNPs) in 208 genes involved in oestrogen and pregnenolone biosynthesis and oestrogen-mediated signalling. In the discovery phase 14 polymorphisms showed a statistically significant association (P < 0.05). In the replication none of the findings were validated. In addition, a gene-based analysis was performed on the 208 selected genes. Four genes (NR5A2, MED1, NCOA2 and RUNX1) were associated with PDAC risk, but only NR5A2 showed an association (P = 4.08 × 10-5) below the Bonferroni-corrected threshold of statistical significance. In conclusion, despite differences in incidence between males and females, our study did not identify an effect of common polymorphisms in the oestrogen and pregnenolone pathways in relation to PDAC susceptibility. However, we validated the previously reported association between NR5A2 gene variants and PDAC risk.
Collapse
Affiliation(s)
- Giulia Peduzzi
- grid.5395.a0000 0004 1757 3729Department of Biology, University of Pisa, 56126 Pisa, Italy
| | - Livia Archibugi
- grid.415230.10000 0004 1757 123XDigestive and Liver Disease Unit, Sant’Andrea Hospital, Rome, Italy ,grid.18887.3e0000000417581884Pancreato-Biliary Endoscopy and Endoscopic Ultrasound, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Verena Katzke
- grid.7497.d0000 0004 0492 0584Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manuel Gentiluomo
- grid.5395.a0000 0004 1757 3729Department of Biology, University of Pisa, 56126 Pisa, Italy
| | - Gabriele Capurso
- grid.415230.10000 0004 1757 123XDigestive and Liver Disease Unit, Sant’Andrea Hospital, Rome, Italy ,grid.18887.3e0000000417581884Pancreato-Biliary Endoscopy and Endoscopic Ultrasound, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | | | - Maria Gazouli
- grid.5216.00000 0001 2155 0800Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Mara Goetz
- grid.13648.380000 0001 2180 3484Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Hermann Brenner
- grid.7497.d0000 0004 0492 0584Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.7497.d0000 0004 0492 0584Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Renata Talar-Wojnarowska
- grid.8267.b0000 0001 2165 3025Department of Digestive Tract Diseases, Medical University of Lodz, Lodz, Poland
| | - Giuseppe Vanella
- grid.415230.10000 0004 1757 123XDigestive and Liver Disease Unit, Sant’Andrea Hospital, Rome, Italy ,grid.18887.3e0000000417581884Pancreato-Biliary Endoscopy and Endoscopic Ultrasound, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Francesca Tavano
- grid.413503.00000 0004 1757 9135Division of Gastroenterology and Research Laboratory, Fondazione IRCCS “Casa Sollievo Della Sofferenza” Hospital, San Giovanni Rotondo, Italy
| | - Maurizio Lucchesi
- Oncology of Massa Carrara, Oncological Department, Azienda USL Toscana Nord Ovest, Carrara, Italy
| | - Beatrice Mohelnikova-Duchonova
- grid.10979.360000 0001 1245 3953Department of Oncology, Faculty of Medicine and Dentistry, Palacký University, Olomouc, Czech Republic
| | - Xuechen Chen
- grid.7497.d0000 0004 0492 0584Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Vytautas Kiudelis
- grid.45083.3a0000 0004 0432 6841Gastroenterology Department and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Péter Hegyi
- grid.9679.10000 0001 0663 9479Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary ,grid.11804.3c0000 0001 0942 9821Centre for Translational Medicine, Semmelweis University, Budapest, Hungary ,grid.11804.3c0000 0001 0942 9821Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, Budapest, Hungary ,grid.9679.10000 0001 0663 9479János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Martin Oliverius
- grid.4491.80000 0004 1937 116XSurgery Clinic Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Hannah Stocker
- grid.7497.d0000 0004 0492 0584Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Network Aging Research (NAR), Heidelberg University, Heidelberg, Germany
| | - Caterina Stornello
- grid.415230.10000 0004 1757 123XDigestive and Liver Disease Unit, Sant’Andrea Hospital, Rome, Italy ,grid.18887.3e0000000417581884Pancreato-Biliary Endoscopy and Endoscopic Ultrasound, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Ludmila Vodickova
- grid.424967.a0000 0004 0404 6946Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic ,grid.4491.80000 0004 1937 116XFirst Faculty of Medicine, Institute of Biology and Medical Genetics, Charles University, Prague, Czech Republic ,grid.4491.80000 0004 1937 116XBiomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Souček
- grid.4491.80000 0004 1937 116XBiomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - John P. Neoptolemos
- grid.7700.00000 0001 2190 4373Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | - Sabrina Gloria Giulia Testoni
- grid.18887.3e0000000417581884Pancreato-Biliary Endoscopy and Endoscopic Ultrasound, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Luca Morelli
- grid.5395.a0000 0004 1757 3729General Surgery, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Rita T. Lawlor
- grid.411475.20000 0004 1756 948XARC-NET, Centre for Applied Research on Cancer, University and Hospital Trust of Verona, Verona, Italy
| | - Daniela Basso
- grid.5608.b0000 0004 1757 3470Department DIMED-Laboratory Medicine, University of Padova, Padua, Italy
| | - Jakob R. Izbicki
- grid.13648.380000 0001 2180 3484Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Stefano Ermini
- grid.413181.e0000 0004 1757 8562Blood Transfusion Service, Azienda Ospedaliero-Universitaria Meyer, Florence, Italy
| | - Juozas Kupcinskas
- grid.45083.3a0000 0004 0432 6841Gastroenterology Department and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | - Ugo Boggi
- grid.144189.10000 0004 1756 8209Division of General and Transplant Surgery, Pisa University Hospital, Pisa, Italy
| | - Hanneke W. M. van Laarhoven
- grid.7177.60000000084992262Department of Medical Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands ,grid.16872.3a0000 0004 0435 165XCancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Andrea Szentesi
- grid.9679.10000 0001 0663 9479Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary ,grid.9679.10000 0001 0663 9479János Szentágothai Research Center, University of Pécs, Pécs, Hungary ,grid.9008.10000 0001 1016 9625Centre for Translational Medicine, Department of Medicine, University of Szeged, Szeged, Hungary
| | - Bálint Erőss
- grid.9679.10000 0001 0663 9479Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary ,grid.11804.3c0000 0001 0942 9821Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, Budapest, Hungary ,grid.11804.3c0000 0001 0942 9821Center for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Giovanni Capretti
- grid.452490.eDepartment of Biomedical Sciences, Humanitas University, Milan, Italy ,grid.417728.f0000 0004 1756 8807Pancreatic Surgery Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Ben Schöttker
- grid.7497.d0000 0004 0492 0584Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jurgita Skieceviciene
- grid.45083.3a0000 0004 0432 6841Gastroenterology Department and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Mateus Nóbrega Aoki
- grid.418068.30000 0001 0723 0931Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Casper H. J. van Eijck
- grid.5645.2000000040459992XDepartment of Surgery, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Giulia Martina Cavestro
- grid.15496.3f0000 0001 0439 0892Gastroenterology and Gastrointestinal Endoscopy Unit, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federico Canzian
- grid.7497.d0000 0004 0492 0584Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniele Campa
- grid.5395.a0000 0004 1757 3729Department of Biology, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
16
|
Dehydroepiandrosterone Sulfate and Colorectal Cancer Risk: A Mendelian Randomization Analysis. Twin Res Hum Genet 2022; 25:180-186. [PMID: 36053043 DOI: 10.1017/thg.2022.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Colorectal cancer is the third most common and second most deadly type of cancer worldwide, with approximately 1.9 million cases and 0.9 million deaths worldwide in 2020. Previous studies have shown that estrogen and testosterone hormones are associated with colorectal cancer risk and mortality. However, the potential effect of their precursor, dehydroepiandrosterone sulfate (DHEAS), on colorectal cancer risk has not been investigated. Therefore, evaluating DHEAS's effect on colorectal cancer will expand our understanding of the hormonal contribution to colorectal cancer risk. In this study, we conducted a two-sample Mendelian randomization (MR) analysis to investigate the causal effect of DHEAS on colorectal cancer. We obtained DHEAS and colorectal cancer genomewide association study (GWAS) summary statistics from the Leipzig Health Atlas and the GWAS catalog and conducted MR analyses using the TwoSampleMR R package. Our results suggest that higher DHEAS levels are causally associated with decreased colorectal cancer risk (odds ratio per unit increase in DHEAS levels z score = 0.70; 95% confidence interval [0.51, 0.96]), which is in line with previous observations in a case-control study of colon cancer. The outcome of this study will be beneficial in developing plasma DHEAS-based biomarkers in colorectal cancer. Further studies should be conducted to interpret the DHEAS-colorectal cancer association among different ancestries and populations.
Collapse
|
17
|
Johansson Å, Schmitz D, Höglund J, Hadizadeh F, Karlsson T, Ek WE. Investigating the Effect of Estradiol Levels on the Risk of Breast, Endometrial, and Ovarian Cancer. J Endocr Soc 2022; 6:bvac100. [PMID: 35822202 PMCID: PMC9265484 DOI: 10.1210/jendso/bvac100] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Indexed: 11/19/2022] Open
Abstract
Background High levels of estrogen are associated with increased risk of breast and endometrial cancer and have been suggested to also play a role in the development of ovarian cancer. Cancerogenic effects of estradiol, the most prominent form of estrogen, have been highlighted as a side effect of estrogen-only menopausal hormone therapy. However, whether high levels of endogenous estrogens, produced within the body, promote cancer development, has not been fully established. Objective We aimed to examine causal effects of estradiol on breast, endometrial, and ovarian cancer. Methods Here we performed a two-sample Mendelian randomization (MR) to estimate the effect of endogenous estradiol on the risk of developing breast, endometrial, and ovarian cancer, using the UK Biobank as well as 3 independent cancer cohorts. Results Using 3 independent instrumental variables, we showed that higher estradiol levels significantly increase the risk for ovarian cancer (OR = 3.18 [95% CI, 1.47-6.87], P = 0.003). We also identified a nominally significant effect for ER-positive breast cancer (OR = 2.16 [95% CI, 1.09-4.26], P = 0.027). However, we could not establish a clear link to the risk of endometrial cancer (OR = 1.93 [95% CI, 0.77-4.80], P = 0.160). Conclusion Our results suggest that high estradiol levels promote the development of ovarian and ER-positive breast cancer.
Collapse
Affiliation(s)
- Åsa Johansson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 75108 Uppsala, Sweden
| | - Daniel Schmitz
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 75108 Uppsala, Sweden
| | - Julia Höglund
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 75108 Uppsala, Sweden
| | - Fatemeh Hadizadeh
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 75108 Uppsala, Sweden
| | - Torgny Karlsson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 75108 Uppsala, Sweden
| | - Weronica E Ek
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 75108 Uppsala, Sweden
| |
Collapse
|
18
|
Molenberg R, Thio CHL, Aalbers MW, Uyttenboogaart M, Larsson SC, Bakker MK, Ruigrok YM, Snieder H, van Dijk JMC. Sex Hormones and Risk of Aneurysmal Subarachnoid Hemorrhage: A Mendelian Randomization Study. Stroke 2022; 53:2870-2875. [PMID: 35652345 PMCID: PMC9389934 DOI: 10.1161/strokeaha.121.038035] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The risk of aneurysmal subarachnoid hemorrhage (aSAH) is increased in postmenopausal women compared with men of similar age, suggesting a role for sex hormones. We aimed to explore whether sex hormones, and age at menarche/menopause have a causal effect on aSAH risk by conducting a 2-sample MR study (Mendelian randomization).
Collapse
Affiliation(s)
- Rob Molenberg
- Department of Neurosurgery (R.M., M.W.A., J.M.C.v.D.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Chris H L Thio
- Department of Epidemiology (C.H.L.T., H.S.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Marlien W Aalbers
- Department of Neurosurgery (R.M., M.W.A., J.M.C.v.D.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Maarten Uyttenboogaart
- Department of Neurology and Medical Imaging Center (M.U.), University of Groningen, University Medical Center Groningen, the Netherlands
| | | | - Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden (S.C.L.).,Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Sweden (S.C.L.)
| | - Mark K Bakker
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, University Utrecht, the Netherlands (M.K.B., Y.M.R.)
| | - Ynte M Ruigrok
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, University Utrecht, the Netherlands (M.K.B., Y.M.R.)
| | - Harold Snieder
- Department of Epidemiology (C.H.L.T., H.S.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - J Marc C van Dijk
- Department of Neurosurgery (R.M., M.W.A., J.M.C.v.D.), University of Groningen, University Medical Center Groningen, the Netherlands
| |
Collapse
|
19
|
Peng H, Wu X, Wen Y. Plasma Circulating Vitamin C Levels and Risk of Endometrial Cancer: A Bi-Directional Mendelian Randomization Analysis. Front Med (Lausanne) 2022; 9:792008. [PMID: 35402429 PMCID: PMC8984247 DOI: 10.3389/fmed.2022.792008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/31/2022] [Indexed: 11/18/2022] Open
Abstract
Background Observational studies indicated that circulating vitamin C (VitC) levels may be correlated with the risk of endometrial cancer (EC). However, the causal effects and direction between them were still unclear. Methods In this study, 11 single nucleotide polymorphisms (SNPs) robustly correlated with plasma VitC levels were extracted from the latest genome-wide association study (GWAS), containing 52,018 individuals. Genetic data of EC were obtained from the Endometrial Cancer Association Consortium (ECAC) (12,906 cases and 108,979 controls). An inverse-variance weighted method was utilized as the primary analysis of Mendelian randomization (MR), supplemented by the weighted median, MR Pleiotropy Residual Sum and Outlier test (MR-PRESSO), and MR-Egger methods. Additional sensitivity analyses excluding 3 SNPs with secondary phenotypes were conducted to rule out the possible pleiotropic effects. Potential impacts of several risk factors of EC, such as obesity, body mass index (BMI), hypertension, and diabetes on VitC levels, were assessed. We additionally evaluated the effects of VitC on LDL cholesterol levels, HDL cholesterol levels, and triglycerides levels to probe into the possible mediators in the VitC-EC pathway. Results Genetically predicted higher plasma VitC levels (per 1 SD increase, approximately 20 μmol/L) were causally associated with an increased risk of EC overall [odds ratio (OR) 1.374, 95% CI 1.128–1.674, p = 0.0016], supported by complementary sensitivity analyses. In the subgroup analyses, genetically predicted higher levels of VitC were associated with a tendency of increased risks of both endometrioid (ORSD 1.324, 95% CI 0.959–1.829, p = 0.0881) and non-endometrioid histology (ORSD 1.392, 95% CI 0.873–2.220, p = 0.1647) while without statistical significance. The association remained significant after the exclusion of the three pleiotropic SNPs (ORSD 1.394, 95% CI 1.090–1.784, p = 0.0082). The confounders and mediators were unlikely to affect the VitC-EC relationship. The causal effect of EC on VitC levels was not supported (OR 1.001, 95% CI 0.998–1.004, p = 0.4468). Conclusions This bi-directional MR study demonstrated a causal risk role of higher circulating VitC at physiological levels on an increased risk of EC, which was independent of confounders and mediators. Further studies are warranted to elucidate the possible mechanisms.
Collapse
Affiliation(s)
- Haoxin Peng
- Nanshan School, Guangzhou Medical University, Guangzhou, China
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Haoxin Peng
| | - Xiangrong Wu
- Nanshan School, Guangzhou Medical University, Guangzhou, China
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yaokai Wen
- Department of Medical Oncology, School of Medicine, Tongji University, Shanghai, China
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Guo JZ, Wu QJ, Liu FH, Gao C, Gong TT, Li G. Review of Mendelian Randomization Studies on Endometrial Cancer. Front Endocrinol (Lausanne) 2022; 13:783150. [PMID: 35615721 PMCID: PMC9124776 DOI: 10.3389/fendo.2022.783150] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/30/2022] [Indexed: 01/04/2023] Open
Abstract
Endometrial cancer (EC) is a common gynecological cancer. In some parts of the world, the incidence and mortality of EC are on the rise. Understanding the risk factors of EC is necessary to prevent the occurrence of this disease. Observational studies have revealed the association between certain modifiable environmental risk factors and EC risk. However, due to unmeasured confounding, measurement errors, and reverse causality, observational studies sometimes have limited ability to judge robust causal inferences. In recent years, Mendelian randomization (MR) analysis has received extensive attention, providing valuable insights for cancer-related research, and is expected to identify potential therapeutic interventions. In MR analysis, genetic variation (alleles are randomly assigned during meiosis and are usually independent of environmental or lifestyle factors) is used instead of modifiable exposure to study the relationship between risk factors and disease. Therefore, MR analysis can make causal inference about exposure and disease risk. This review briefly describes the key principles and assumptions of MR analysis; summarizes published MR studies on EC; focuses on the correlation between different risk factors and EC risks; and discusses the application of MR methods in EC research. The results of MR studies on EC showed that type 2 diabetes, uterine fibroids, higher body mass index, higher plasminogen activator inhibitor-1 (PAI-1), higher fasting insulin, early insulin secretion, longer telomere length, higher testosterone and higher plasma cortisol levels are associated with increased risk of EC. In contrast, later age of menarche, higher circulatory tumor necrosis factor, higher low-density lipoprotein cholesterol, and higher sex hormone-binding globulin levels are associated with reduced risk of EC. In general, despite some limitations, MR analysis still provides an effective way to explore the causal relationship between different risk factors and EC.
Collapse
Affiliation(s)
- Jian-Zeng Guo
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qi-Jun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fang-Hua Liu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chang Gao
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ting-Ting Gong
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Gang Li, ; Ting-Ting Gong,
| | - Gang Li
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Gang Li, ; Ting-Ting Gong,
| |
Collapse
|
21
|
Bogush TA, Basharina AA, Bogush EA, Scherbakov AM, Davydov MM, Kosorukov VS. The expression and clinical significance of ERβ/ERα in ovarian cancer: can we predict the effectiveness of platinum plus taxane therapy? Ir J Med Sci 2021; 191:2047-2053. [PMID: 34741718 DOI: 10.1007/s11845-021-02842-6] [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: 10/06/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022]
Abstract
Estrogens play an extremely important role in regulating the proliferation of ovarian cancer. The estrogen receptor alpha (ERα) stimulates cell growth, whereas ERβ can be attributed to tumor suppressors. The study aims to assess the relationship between the expression of estrogen receptors in tumors and the efficacy of front-line platinum plus taxane chemotherapy in ovarian cancer patients. MATERIALS AND METHODS ERα and ERβ tumor expression was evaluated quantitatively by flow cytometry in a narrowly defined group (31 patients): stage III high-grade serous ovarian carcinoma (HGSOC), suboptimal surgical cytoreduction, front-line platinum plus taxane chemotherapy (front-line, six cycles). RESULTS The median of progression-free survival (PFS) was 2 times greater (18 vs 8 months, p = 0.04) and the recurrence risk (HR) was 2.2 times (95 % CI: 1.1-6.2, p = 0.04) lower in the group with high (in more than 40% of the cells) vs low level of ERβ tumor expression. The statistically significant difference between PFS in the groups with high vs low tumor ERα expression was not revealed. CONCLUSION A high level of ERβ and not ERα expression can predict the efficacy of front-line platinum plus taxane chemotherapy in stage III HGSOC patients. The status of estrogen receptor beta can be considered as one of the possible predictors for evaluating the effectiveness of ovarian cancer therapy.
Collapse
Affiliation(s)
- Tatiana A Bogush
- Group of Molecular Tumor Markers, N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - Anna A Basharina
- Group of Molecular Tumor Markers, N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - Elena A Bogush
- Department of Oncology, N.V. Sklifosovsky Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russian Federation
| | - Alexander M Scherbakov
- Laboratory of Oncoproteomics, N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, Moscow, Russian Federation.
| | - Mikhail M Davydov
- Department of Oncology, N.V. Sklifosovsky Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russian Federation
| | - Vyacheslav S Kosorukov
- Laboratory of Transgenic Drugs, N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, Moscow, Russian Federation
| |
Collapse
|