1
|
Zhu P, Ren J, Sun J, Geng J, Wang H, Ma M. The association of endogenous sex hormones with endometrial cancer risk: A systematic review and meta-analysis. Eur J Obstet Gynecol Reprod Biol 2025; 310:113997. [PMID: 40300286 DOI: 10.1016/j.ejogrb.2025.113997] [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: 12/29/2024] [Revised: 04/20/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025]
Abstract
OBJECTIVES The role of endogenous sex hormones in the risk of endometrial cancer (EC) remains contradictory across the studies. This meta-analysis was carried out to investigate the relation of circulating concentrations of sex hormones and sex hormone-binding globulin (SHBG) to EC risk. METHODS A search of the PubMed, Web of Science, and Scopus databases was conducted to include relevant studies. We used odds ratios (OR) with 95% confidence intervals (CI) to pool effect sizes using a random effects model. RESULTS The analysis included 16 studies with 292,695 participants. SHBG levels showed an inverse relationship with EC (OR: 0.67). In contrast, higher circulating levels of total testosterone (OR: 1.70), free testosterone (OR: 1.75), dehydroepiandrosterone sulfate (OR: 1.39), and androstenedione (OR: 1.58) were positively associated with EC risk. Estrogens also demonstrated significant associations, so that estrone (OR: 1.55), unconjugated estrone (OR: 1.86), estradiol (OR: 1.38), unconjugated estradiol (OR: 2.14), estriol (OR: 1.75), and unconjugated estriol (OR: 1.99) were linked to increased EC risk, while conjugated estrogens showed no significant associations. A non-linear dose-response relationship was found for SHBG, estrone, estradiol, and total testosterone. The results were significantly affected by age, cancer type, geographic region, menopausal status, study type, and the level of adjustments for covariates. For all hormones, the significant associations were found only for postmenopausal women. CONCLUSIONS This study found an inverse association between SHBG and EC, while identified a direct relationship between sex hormones, except for conjugated estrogens, and EC risk only in postmenopausal women.
Collapse
Affiliation(s)
- Pengfei Zhu
- Department of Gynecology, Dezhou Women and Children's Hospital, Dezhou 253000, China
| | - Juanjuan Ren
- Department of Gynecology, Dezhou Women and Children's Hospital, Dezhou 253000, China
| | - Jing Sun
- Department of Gynecology, Dezhou Women and Children's Hospital, Dezhou 253000, China
| | - Jingjing Geng
- Department of Gynecology, Dezhou Women and Children's Hospital, Dezhou 253000, China
| | - Huan Wang
- Department of Gynecology, Dezhou Women and Children's Hospital, Dezhou 253000, China
| | - Mingming Ma
- Department of Gynecology, Dezhou Women and Children's Hospital, Dezhou 253000, China.
| |
Collapse
|
2
|
Park K, Shin CM, Kim N, Won S, Song CH, Ohn JH, Lee S, Park JH, Yie GE, Kang SJ, Kim JS, Lee DH. rs762855 single nucleotide polymorphism modulates the risk for diffuse-type gastric cancer in females: a genome-wide association study in the Korean population. Gastric Cancer 2025; 28:145-159. [PMID: 39862296 PMCID: PMC11842433 DOI: 10.1007/s10120-024-01575-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 12/08/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Intestinal-type gastric cancer (IGC) and diffuse-type gastric cancer (DGC) exhibit different prevalence rates between sexes. While environmental factors like Helicobacter pylori infection and alcohol consumption contribute to these differences, they do not fully account for them, suggesting a role for host genetic factors. METHODS We conducted a meta-analysis to explore associations between single nucleotide polymorphisms (SNPs) and the risk of IGC or DGC. The analysis included the SNUBH cohort (998 participants: 159 DGCs, 303 IGCs, 4,962,361 variants) and the GC_HC cohort (6,233 participants: 389 DGCs, 405 IGCs, 4,541,617 variants). Significant variants were validated in the SNUBH2_AA cohort (5,511 participants: 40 DGCs, 49 IGCs, 3,668,632 variants). RESULTS The meta-analysis identified that rs762855 (chr4:3,074,795; hg19) is significantly associated with DGC risk in females (OR [95% CI]: 1.758 [1.438-2.150], P = 3.91 × 10-8), a finding replicated in the SNUBH2_AA datasets (OR [95% CI]: 3.356 [1.031-10.92], P = 4.43 × 10-2). Gene-set and transcriptomic analyses revealed that the Myb/SANT DNA Binding Domain Containing 1 (MSANTD1) gene is significantly linked to DGC susceptibility in females. In addition, Mendelian randomization analyses suggested that increased serum total protein and non-albumin protein (NAP) levels elevate DGC risk in females (P < 0.05), but not in males. CONCLUSION The rs762855 SNP, MSANTD1, and serum NAP levels are associated with DGC risk in Korean females.
Collapse
Affiliation(s)
- Kyungtaek Park
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-Do, South Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-Do, South Korea.
- Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, 173-82, Gumi-ro, Bundang-gu, Seongnam, Gyeonggi-Do, South Korea.
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
| | - Sungho Won
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
- Department of Public Health Sciences, Seoul National University, Seoul, South Korea
- Interdisciplinary Program of Bioinformatics, Seoul National University, Seoul, Korea
| | - Chin-Hee Song
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-Do, South Korea
| | - Jung Hun Ohn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-Do, South Korea
| | - Sejoon Lee
- Precision Medicine Center, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Ji Hyun Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Ga-Eun Yie
- Interdisciplinary Program of Bioinformatics, Seoul National University, Seoul, Korea
| | - Seung Joo Kang
- Department of Internal Medicine and Healthcare Research Institute, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Korea
| | - Joo Sung Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Department of Internal Medicine and Healthcare Research Institute, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-Do, South Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
3
|
Lu XF, Huang T, Chen C, Zhang J, Fu XY, Cheng B, Zhou YY, Lei J, Lu DL. Association of CYP7B1 expression with the prognosis of endometrial cancer: a retrospective study. World J Surg Oncol 2024; 22:251. [PMID: 39289693 PMCID: PMC11406946 DOI: 10.1186/s12957-024-03504-4] [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: 02/09/2024] [Accepted: 08/13/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Endometrial cancer (EC) tissues express CYP7B1, but its association with prognosis needs to be investigated. METHODS Immunohistochemistry and image analysis software were used to assess CYP7B1 protein expression in paraffin-embedded endometrial tumor sections. Associations between CYP7B1 and clinical factors were tested with the Wilcoxon rank-sum test. Kaplan-Meier curves were employed to describe survival, and differences were assessed using the log-rank test. Cox regression analysis was used to assess the association between CYP7B1 expression and the prognosis of patients with EC. RESULTS A total of 307 patients were enrolled with an average age of 52.6 ± 8.0 years at diagnosis. During the period of follow-up, 46 patients (15.0%) died, and 29 (9.4%) suffered recurrence. The expression of CYP7B1 protein is significantly higher in the cytoplasm than in the nucleus (P < 0.001). Patients aged < 55 years (P = 0.040), ER-positive patients (P = 0.028) and PR-positive patients (P < 0.001) report higher levels of CYP7B1 protein. Both univariate (HR = 0.41, 95% CI: 0.18-0.90, P = 0.025) and multivariate (HR = 0.35, 95%CI:0.16-0.79, P = 0.011) Cox regression analyses demonstrate that high CYP7B1 protein expression predicts longer overall survival (OS). When considering only ER-positive patients (n = 265), CYP7B1 protein expression is more strongly associated with OS (HR = 0.20,95%CI:0.08-0.52, P = 0.001). The 3-year OS and 5-year OS in the low-CYP7B1 subgroup are 81.6% and 76.8%, respectively; while in the high-CYP7B1 subgroup are 93.0% and 92.0%, respectively (P = 0.021). CONCLUSIONS High CYP7B1 protein expression predicted longer OS, suggesting that it may serve as an important molecular marker for EC prognosis.
Collapse
Affiliation(s)
- Xiao-Fang Lu
- Department of Epidemiology, School of Medicine, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| | - Tao Huang
- Department of Gynecology, Wuzhou Red Cross Hospital, No. 3-1, Xinxing 1st Road, Wanxiu District, Wuzhou, 543002, China
| | - Chang Chen
- Department of Pathology, Wuzhou Red Cross Hospital, Wuzhou, China
| | - Jing Zhang
- Department of Epidemiology, School of Medicine, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| | - Xu-Yong Fu
- Department of Epidemiology, School of Medicine, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| | - Bo Cheng
- Department of Pathology, Chinese People's Liberation Army Rocket Force Characteristic Medical Center, Beijing, China
| | - Ya-Yan Zhou
- Department of Radiation Oncology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Jia Lei
- Department of Gynecology, Wuzhou Red Cross Hospital, No. 3-1, Xinxing 1st Road, Wanxiu District, Wuzhou, 543002, China.
| | - Da-Lin Lu
- Department of Epidemiology, School of Medicine, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China.
| |
Collapse
|
4
|
Bostan IS, Mihaila M, Roman V, Radu N, Neagu MT, Bostan M, Mehedintu C. Landscape of Endometrial Cancer: Molecular Mechanisms, Biomarkers, and Target Therapy. Cancers (Basel) 2024; 16:2027. [PMID: 38893147 PMCID: PMC11171255 DOI: 10.3390/cancers16112027] [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: 04/15/2024] [Revised: 05/20/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Endometrial cancer is one the most prevalent gynecological cancers and, unfortunately, has a poor prognosis due to low response rates to traditional treatments. However, the progress in molecular biology and understanding the genetic mechanisms involved in tumor processes offers valuable information that has led to the current classification that describes four molecular subtypes of endometrial cancer. This review focuses on the molecular mechanisms involved in the pathogenesis of endometrial cancers, such as genetic mutations, defects in the DNA mismatch repair pathway, epigenetic changes, or dysregulation in angiogenic or hormonal signaling pathways. The preclinical genomic and molecular investigations presented allowed for the identification of some molecules that could be used as biomarkers to diagnose, predict, and monitor the progression of endometrial cancer. Besides the therapies known in clinical practice, targeted therapy is described as a new cancer treatment that involves identifying specific molecular targets in tumor cells. By selectively inhibiting these targets, key signaling pathways involved in cancer progression can be disrupted while normal cells are protected. The connection between molecular biomarkers and targeted therapy is vital in the fight against cancer. Ongoing research and clinical trials are exploring the use of standard therapy agents in combination with other treatment strategies like immunotherapy and anti-angiogenesis therapy to improve outcomes and personalize treatment for patients with endometrial cancer. This approach has the potential to transform the management of cancer patients. In conclusion, enhancing molecular tools is essential for stratifying the risk and guiding surgery, adjuvant therapy, and cancer treatment for women with endometrial cancer. In addition, the information from this review may have an essential value in the personalized therapy approach for endometrial cancer to improve the patient's life.
Collapse
Affiliation(s)
| | - Mirela Mihaila
- Stefan S. Nicolau Institute of Virology, Center of Immunology, Romanian Academy, 030304 Bucharest, Romania; (M.M.); (V.R.)
- Faculty of Pharmacy, Titu Maiorescu University, 040314 Bucharest, Romania
| | - Viviana Roman
- Stefan S. Nicolau Institute of Virology, Center of Immunology, Romanian Academy, 030304 Bucharest, Romania; (M.M.); (V.R.)
| | - Nicoleta Radu
- Department of Biotechnology, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 011464 Bucharest, Romania;
- Biotechnology Department, National Institute for Chemistry and Petrochemistry R&D of Bucharest, 060021 Bucharest, Romania
| | - Monica Teodora Neagu
- Department of Immunology, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania;
| | - Marinela Bostan
- Stefan S. Nicolau Institute of Virology, Center of Immunology, Romanian Academy, 030304 Bucharest, Romania; (M.M.); (V.R.)
- Department of Immunology, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania;
| | - Claudia Mehedintu
- Filantropia Clinical Hospital, 011132 Bucharest, Romania; (I.-S.B.); (C.M.)
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, 050471 Bucharest, Romania
| |
Collapse
|
5
|
Bukato K, Kostrzewa T, Gammazza AM, Gorska-Ponikowska M, Sawicki S. Endogenous estrogen metabolites as oxidative stress mediators and endometrial cancer biomarkers. Cell Commun Signal 2024; 22:205. [PMID: 38566107 PMCID: PMC10985914 DOI: 10.1186/s12964-024-01583-0] [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: 11/29/2023] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Endometrial cancer is the most common gynecologic malignancy found in developed countries. Because therapy can be curative at first, early detection and diagnosis are crucial for successful treatment. Early diagnosis allows patients to avoid radical therapies and offers conservative management options. There are currently no proven biomarkers that predict the risk of disease occurrence, enable early identification or support prognostic evaluation. Consequently, there is increasing interest in discovering sensitive and specific biomarkers for the detection of endometrial cancer using noninvasive approaches. CONTENT Hormonal imbalance caused by unopposed estrogen affects the expression of genes involved in cell proliferation and apoptosis, which can lead to uncontrolled cell growth and carcinogenesis. In addition, due to their ability to cause oxidative stress, estradiol metabolites have both carcinogenic and anticarcinogenic properties. Catechol estrogens are converted to reactive quinones, resulting in oxidative DNA damage that can initiate the carcinogenic process. The molecular anticancer mechanisms are still not fully understood, but it has been established that some estradiol metabolites generate reactive oxygen species and reactive nitrogen species, resulting in nitro-oxidative stress that causes cancer cell cycle arrest or cell death. Therefore, identifying biomarkers that reflect this hormonal imbalance and the presence of endometrial cancer in minimally invasive or noninvasive samples such as blood or urine could significantly improve early detection and treatment outcomes.
Collapse
Affiliation(s)
- Katarzyna Bukato
- Department of Obstetrics and Gynecology, Oncological Gynecology and Gynecological Endocrinology, Medical University of Gdansk, Smoluchowskiego 17, Gdańsk, 80-214, Poland
| | - Tomasz Kostrzewa
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, Dębinki 1, Gdansk, 80-211, Poland
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, 90127, Italy
| | - Magdalena Gorska-Ponikowska
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, Dębinki 1, Gdansk, 80-211, Poland.
- IEMEST Istituto Euro-Mediterraneo di Scienza e Tecnologia, Palermo, 90127, Italy.
- Department of Biophysics, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, 70174, Stuttgart, Germany.
| | - Sambor Sawicki
- Department of Obstetrics and Gynecology, Oncological Gynecology and Gynecological Endocrinology, Medical University of Gdansk, Smoluchowskiego 17, Gdańsk, 80-214, Poland.
| |
Collapse
|
6
|
Zhao Y, Tan H, Zhang J, Zhan D, Yang B, Hong S, Pan B, Wang N, Chen T, Shi Y, Wang Z. Developing liver-targeted naringenin nanoparticles for breast cancer endocrine therapy by promoting estrogen metabolism. J Nanobiotechnology 2024; 22:122. [PMID: 38504208 PMCID: PMC10953142 DOI: 10.1186/s12951-024-02356-0] [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: 12/31/2023] [Accepted: 02/20/2024] [Indexed: 03/21/2024] Open
Abstract
Endocrine therapy is standard for hormone receptor-positive (HR+) breast cancer treatment. However, current strategies targeting estrogen signaling pay little attention to estradiol metabolism in the liver and is usually challenged by treatment failure. In a previous study, we demonstrated that the natural compound naringenin (NAR) inhibited HR+ breast cancer growth by activating estrogen sulfotransferase (EST) expression in the liver. Nevertheless, the poor water solubility, low bio-barrier permeability, and non-specific distribution limited its clinical application, particularly for oral administration. Here, a novel nano endocrine drug NAR-cell penetrating peptide-galactose nanoparticles (NCG) is reported. We demonstrated that NCG presented specific liver targeting and increased intestinal barrier permeability in both cell and zebrafish xenotransplantation models. Furthermore, NCG showed liver targeting and enterohepatic circulation in mouse breast cancer xenografts following oral administration. Notably, the cancer inhibition efficacy of NCG was superior to that of both NAR and the positive control tamoxifen, and was accompanied by increased hepatic EST expression and reduced estradiol levels in the liver, blood, and tumor tissue. Moreover, few side effects were observed after NCG treatment. Our findings reveal NCG as a promising candidate for endocrine therapy and highlight hepatic EST targeting as a novel therapeutic strategy for HR+ breast cancer.
Collapse
Affiliation(s)
- Yuying Zhao
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hanxu Tan
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Juping Zhang
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Dandan Zhan
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Bowen Yang
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shicui Hong
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Bo Pan
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Neng Wang
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Yafei Shi
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Zhiyu Wang
- State Key Laboratory of Dampness, Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Clinical Research On Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, China.
| |
Collapse
|
7
|
Petrick JL, Joslin CE, Johnson CE, Camacho TF, Peres LC, Bandera EV, Barnard ME, Beeghly A, Bethea TN, Dempsey LF, Guertin K, Harris HR, Moorman PG, Myers ER, Ochs-Balcom HM, Rosenow W, Setiawan VW, Wu AH, Schildkraut JM, Rosenberg L. Menopausal hormone therapy use and risk of ovarian cancer by race: the ovarian cancer in women of African ancestry consortium. Br J Cancer 2023; 129:1956-1967. [PMID: 37865688 PMCID: PMC10703895 DOI: 10.1038/s41416-023-02407-7] [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: 11/28/2022] [Revised: 08/01/2023] [Accepted: 08/17/2023] [Indexed: 10/23/2023] Open
Abstract
BACKGROUND Most studies examining post-menopausal menopausal hormone therapy (MHT) use and ovarian cancer risk have focused on White women and few have included Black women. METHODS We evaluated MHT use and ovarian cancer risk in Black (n = 800 cases, 1783 controls) and White women (n = 2710 cases, 8556 controls), using data from the Ovarian Cancer in Women of African Ancestry consortium. Logistic regression was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs) for the association of MHT use with ovarian cancer risk, examining histotype, MHT type and duration of use. RESULTS Long-term MHT use, ≥10 years, was associated with an increased ovarian cancer risk for White women (OR = 1.38, 95%CI: 1.22-1.57) and the association was consistent for Black women (OR = 1.20, 95%CI: 0.81-1.78, pinteraction = 0.4). For White women, the associations between long-term unopposed estrogen or estrogen plus progesterone use and ovarian cancer risk were similar; the increased risk associated with long-term MHT use was confined to high-grade serous and endometroid tumors. Based on smaller numbers for Black women, the increased ovarian cancer risk associated with long-term MHT use was apparent for unopposed estrogen use and was predominately confined to other epithelial histotypes. CONCLUSION The association between long-term MHT use and ovarian cancer risk was consistent for Black and White women.
Collapse
Affiliation(s)
| | - Charlotte E Joslin
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago School of Medicine, Chicago, IL, USA
- Division of Epidemiology and Biostatistics, University of Illinois at Chicago School of Public Health, Chicago, IL, USA
| | - Courtney E Johnson
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - T Fabian Camacho
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Lauren C Peres
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Elisa V Bandera
- Cancer Epidemiology and Health Outcomes, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | | | - Alicia Beeghly
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Traci N Bethea
- Office of Minority Health and Health Disparities Research, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Campus, Washington, DC, USA
| | - Lauren F Dempsey
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Kristin Guertin
- Department of Public Health Sciences, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Holly R Harris
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Patricia G Moorman
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Evan R Myers
- Department of Family Medicine and Community Health, Duke University Medical Center, Durham, NC, USA
| | - Heather M Ochs-Balcom
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Will Rosenow
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - V Wendy Setiawan
- University of Southern California Norris Comprehensive Cancer Center and Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Anna H Wu
- University of Southern California Norris Comprehensive Cancer Center and Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Joellen M Schildkraut
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Lynn Rosenberg
- Slone Epidemiology Center at Boston University, Boston, MA, USA
| |
Collapse
|
8
|
Yang S, Zeng Q, Huang X, Liang Z, Hu H. Effect of Isoflavones on Blood Lipid Alterations in Postmenopausal Females: A Systematic Review and Meta-Analysis of Randomized Trials. Adv Nutr 2023; 14:1633-1643. [PMID: 37758058 PMCID: PMC10721517 DOI: 10.1016/j.advnut.2023.09.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/09/2023] [Accepted: 09/21/2023] [Indexed: 10/03/2023] Open
Abstract
The effects of isoflavones on postmenopausal female's blood lipid profile have yielded conflicting results in previous studies. Further investigation is necessary to determine the potential benefits of isoflavone therapy in managing cardiovascular health in this population. This meta-analysis aimed to assess the effects of isoflavones on blood lipid concentrations in postmenopausal females. A comprehensive search was conducted in major databases for randomized controlled trials published between 2000 and 2023. Eighteen studies were included in the analysis, which examined the impact of isoflavone intake on blood lipids in postmenopausal females. Isoflavone consumption resulted in a significant reduction in triacylglycerol (TG) concentrations (-12.50 mg/dL; 95% CI: -23.09, -1.91) and a modest increase in high-density lipoprotein cholesterol (HDL cholesterol) concentrations (1.83 mg/dL; 95% CI: 0.03, 3.64). Subgroup analysis showed that isoflavones significantly decreased TG (-15.79 mg/dL; 95% CI: -28.36, -3.22) and increased HDL cholesterol (2.49 mg/dL; 95% CI: 1.80, 3.19) in postmenopausal females under 65 y old. No significant effects were observed in females over 65 y old. Both low (≤80 mg/d) and high (>80 mg/d) doses of isoflavones exhibited TG-lowering effects, whereas only the high dose increased HDL cholesterol. Longer treatment duration (≥24 wk) was associated with a significant reduction in TG, whereas HDL cholesterol improvement occurred during the early period (<24 wk) of supplementation. The consumption of isoflavones resulted in a significant reduction in TG concentrations and an increase in HDL cholesterol concentrations among postmenopausal females under 65 y of age.
Collapse
Affiliation(s)
- Shengmin Yang
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Qingjia Zeng
- Institute of Medical Information/Medical Library, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Xiaohong Huang
- Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhen Liang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| | - Hongpu Hu
- Institute of Medical Information/Medical Library, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China.
| |
Collapse
|
9
|
Gonzalez Deniselle MC, Bettini M, Garrido RM, Meyer M, Lara A, Garay LI, Casas S, Fulgenzi E, Nuñez M, Rugiero MF, De Nicola AF, Gargiulo-Monachelli G. Exposure to endogenous and exogenous sex hormones and reproductive history influence prognosis in women with ALS. Muscle Nerve 2023; 68:414-421. [PMID: 37493444 DOI: 10.1002/mus.27942] [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: 01/28/2023] [Revised: 07/03/2023] [Accepted: 07/08/2023] [Indexed: 07/27/2023]
Abstract
INTRODUCTION/AIMS Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with a higher incidence in men suggesting an influence of sex steroids. Our objective was to investigate past exposure to endogenous and synthetic steroids in female ALS patients and controls. METHODS We administered a questionnaire to 158 postmenopausal women (75 ALS patients and 83 controls). We calculated reproductive time span (RTS), lifetime endogenous estrogen (LEE) and progesterone exposures (LPE), oral contraceptive pill (OCP) use, and reproductive history. RESULTS ALS patients showed shorter LEE and LPE, a lower proportion of breast cancer, and 11% showed no history of pregnancies vs. 4% of controls. Odds ratios (ORs) showed that <17 y of LEE and a delayed menarche (>13 y) constitute risk factors for ALS [OR = 2.1 (95% confidence interval {CI}, 1.08-4.2); and OR = 2.4 (95% CI, 1.1-5.1) respectively]. According to Cox survival analysis, for each year the LEE increased over 17 y, it was independently associated with longer survival [hazard ratio (HR) = 0.37 (95% CI, 0.16-0.85)] after adjusting for smoking, age and site of onset. Multivariate regression analysis demonstrated that for each month using OCP for longer than 40 mo increased the risk of ALS [adjusted OR = 4.1 (95% CI, 1.2-13.8)]. DISCUSSION Thus, longer exposure to endogenous female sex steroids increased survival and reduced ALS susceptibility. In contrast, longer exposure to synthetic sex steroids showed a negative impact by reducing the production of endogenous female sex steroids or due to crossover with other steroid receptors. Given the neuroprotective effects of sex steroids, we suggest that abnormalities of neuroendocrine components may alter motor function in women with ALS.
Collapse
Affiliation(s)
- M Claudia Gonzalez Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Buenos Aires, Argentina
- Department of Physiological Sciences, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Mariela Bettini
- Department of Neurology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Rosa M Garrido
- Department of Gynecology, Hospital Marie Curie, Buenos Aires, Argentina
| | - Maria Meyer
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Buenos Aires, Argentina
| | - Agustina Lara
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Buenos Aires, Argentina
| | - Laura I Garay
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Buenos Aires, Argentina
- Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Sebastian Casas
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Buenos Aires, Argentina
- Department of Neurology, Hospital Militar Central Cirujano Mayor Dr. Cosme Argerich, Buenos Aires, Argentina
| | - Ernesto Fulgenzi
- Hospital General de Agudos Dr. Ignacio Pirovano, Buenos Aires, Argentina
| | - Myriam Nuñez
- Department of Physics Mathematics, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Marcelo F Rugiero
- Department of Gynecology, Hospital Marie Curie, Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Buenos Aires, Argentina
- Department of Neurology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Gisella Gargiulo-Monachelli
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Buenos Aires, Argentina
- Department of Neurology, Hospital General de Agudos Juan A. Fernández, Buenos Aires, Argentina
- Department of Neurology, Centro de Educación Médica e Investigaciones Clínicas "Norberto Quirno", CEMIC, Buenos Aires, Argentina
| |
Collapse
|
10
|
Shah S, Mahamat-Saleh Y, Hajji-Louati M, Correia E, Oulhote Y, Boutron-Ruault MC, Laouali N. Palaeolithic diet score and risk of breast cancer among postmenopausal women overall and by hormone receptor and histologic subtypes. Eur J Clin Nutr 2023; 77:596-602. [PMID: 36726032 DOI: 10.1038/s41430-023-01267-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 02/03/2023]
Abstract
BACKGROUND The Palaeolithic diet (PD) has gained popularity globally. There is emerging evidence of its putative health benefits as short-term effects on chronic diseases have been reported. We evaluated the association between long-term adherence to the PD and breast cancer (BC) risk among postmenopausal women. METHODS 65,574 women from the Etude Epidémiologique auprès de femmes de la Mutuelle Générale de l'Education Nationale (E3N) cohort were followed from 1993 to 2014. Incident BC cases were identified and validated. The PD score was calculated using dietary intake self-reported at baseline (1993) and follow-up (2005) or baseline only if censored before follow-up. Multivariable Cox proportional hazards regression models were used to estimate BC hazard ratios (HR) and 95% confidence intervals (CI). RESULTS Over a mean follow-up of 20 years, 3968 incident BC cases occurred. An increase of 1 standard deviation in the PD score was associated with an 8% lower BC risk, fully-adjusted model: HR1-SD 0.92, 95% CI; 0.89, 0.95. Compared to women with low adherence to the PD, women with high adherence had a 17% lower BC risk, HRQ5 vs Q1 0.83, 95% CI; 0.75, 0.92, Ptrend < 0.01. When considering BC subtypes, we observed the same pattern of association (Pheterogeneity > 0.10 for all). CONCLUSIONS High adherence to a PD characterised by fruit, vegetables, nuts, fish, and lean meat and limited in dairy, grains, legumes, refined sugar, and alcohol was associated with a lower BC risk. The lack of heterogeneity according to BC subtypes could indicate the involvement of non-hormonal mechanisms. The protocol is registered at clinicaltrials.gov as NCT03285230. REGISTRY The protocol is registered at clinicaltrials.gov as NCT03285230.
Collapse
Affiliation(s)
- Sanam Shah
- Paris-Saclay University, UVSQ, Univ. Paris-Sud, Inserm, Gustave Roussy, "Exposome and Heredity" team, CESP, F-94805, Villejuif, France
| | - Yahya Mahamat-Saleh
- International Agency for Research on Cancer (IARC/WHO), 150 cours Albert Thomas, 69372, CEDEX 08, Lyon, France
| | - Mariem Hajji-Louati
- Paris-Saclay University, UVSQ, Univ. Paris-Sud, Inserm, Gustave Roussy, "Exposome and Heredity" team, CESP, F-94805, Villejuif, France
| | - Emmanuelle Correia
- Paris-Saclay University, UVSQ, Univ. Paris-Sud, Inserm, Gustave Roussy, "Exposome and Heredity" team, CESP, F-94805, Villejuif, France
| | - Youssef Oulhote
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, USA
| | - Marie-Christine Boutron-Ruault
- Paris-Saclay University, UVSQ, Univ. Paris-Sud, Inserm, Gustave Roussy, "Exposome and Heredity" team, CESP, F-94805, Villejuif, France.
| | - Nasser Laouali
- Paris-Saclay University, UVSQ, Univ. Paris-Sud, Inserm, Gustave Roussy, "Exposome and Heredity" team, CESP, F-94805, Villejuif, France
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, USA
- Scripps Institution of Oceanography, University of California, San Diego, CA, USA
| |
Collapse
|
11
|
Citrullination: A modification important in the pathogenesis of autoimmune diseases. Clin Immunol 2022; 245:109134. [DOI: 10.1016/j.clim.2022.109134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022]
|
12
|
Le N, Cregger M, Brown V, Loret de Mola J, Bremer P, Nguyen L, Groesch K, Wilson T, Diaz-Sylvester P, Braundmeier-Fleming A. Association of microbial dynamics with urinary estrogens and estrogen metabolites in patients with endometriosis. PLoS One 2021; 16:e0261362. [PMID: 34914785 PMCID: PMC8675749 DOI: 10.1371/journal.pone.0261362] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 11/29/2021] [Indexed: 01/04/2023] Open
Abstract
Endometriosis is an estrogen dependent gynecological disease associated with altered microbial phenotypes. The association among endogenous estrogen, estrogen metabolites, and microbial dynamics on disease pathogenesis has not been fully investigated. Here, we identified estrogen metabolites as well as microbial phenotypes in non-diseased patients (n = 9) and those with pathologically confirmed endometriosis (P-EOSIS, n = 20), on day of surgery (DOS) and ~1–3 weeks post-surgical intervention (PSI). Then, we examined the effects of surgical intervention with or without hormonal therapy (OCPs) on estrogen and microbial profiles of both study groups. For estrogen metabolism analysis, liquid chromatography/tandem mass spectrometry was used to quantify urinary estrogens. The microbiome data assessment was performed with Next generation sequencing to V4 region of 16S rRNA. Surgical intervention and hormonal therapy altered gastrointestinal (GI), urogenital (UG) microbiomes, urinary estrogen and estrogen metabolite levels in P-EOSIS. At DOS, 17β-estradiol was enhanced in P-EOSIS treated with OCPs. At PSI, 16-keto-17β-estradiol was increased in P-EOSIS not receiving OCPs while 2-hydroxyestradiol and 2-hydroxyestrone were decreased in P-EOSIS receiving OCPs. GI bacterial α-diversity was greater for controls and P-EOSIS that did not receive OCPs. P-EOSIS not utilizing OCPs exhibited a decrease in UG bacterial α-diversity and differences in dominant taxa, while P-EOSIS utilizing OCPs had an increase in UG bacterial α-diversity. P-EOSIS had a strong positive correlation between the GI/UG bacteria species and the concentrations of urinary estrogen and its metabolites. These results indicate an association between microbial dysbiosis and altered urinary estrogens in P-EOSIS, which may impact disease progression.
Collapse
Affiliation(s)
- Nhung Le
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
| | - Melissa Cregger
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee, United States of America
- Department of Ecology and Evolutionary Biology, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Veronica Brown
- Division of Biology, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Julio Loret de Mola
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
- Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
| | - Pamela Bremer
- Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
| | - Lyn Nguyen
- Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
| | - Kathleen Groesch
- Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
- Center for Clinical Research, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
| | - Teresa Wilson
- Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
- Center for Clinical Research, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
| | - Paula Diaz-Sylvester
- Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
- Center for Clinical Research, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
| | - Andrea Braundmeier-Fleming
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
- Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
- * E-mail:
| |
Collapse
|
13
|
Gjorgoska M, Rižner TL. Estrogens and the Schrödinger's Cat in the Ovarian Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13195011. [PMID: 34638494 PMCID: PMC8508344 DOI: 10.3390/cancers13195011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/29/2021] [Accepted: 10/02/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Ovarian cancer is a complex pathology for which we require effective screening and therapeutical strategies. Apart from the cancer cell portion, there exist plastic immune and non-immune cell populations, jointly constituting the context-adaptive tumor microenvironment, which is pivotal in tumorigenesis. Estrogens might be synthesized in the ovarian tumor tissue and actively contribute to the shaping of an immunosuppressive microenvironment. Current immune therapies have limited effectiveness as a multitude of factors influence the outcome. A thorough understanding of the ovarian cancer biology is crucial in the efforts to reestablish homeostasis. Abstract Ovarian cancer is a heterogeneous disease affecting the aging ovary, in concert with a complex network of cells and signals, together representing the ovarian tumor microenvironment. As in the “Schrödinger’s cat” thought experiment, the context-dependent constituents of the—by the time of diagnosis—well-established tumor microenvironment may display a tumor-protective and -destructive role. Systemic and locally synthesized estrogens contribute to the formation of a pro-tumoral microenvironment that enables the sustained tumor growth, invasion and metastasis. Here we focus on the estrogen biosynthetic and metabolic pathways in ovarian cancer and elaborate their actions on phenotypically plastic, estrogen-responsive, aging immune cells of the tumor microenvironment, altogether highlighting the multicomponent-connectedness and complexity of cancer, and contributing to a broader understanding of the ovarian cancer biology.
Collapse
|
14
|
Trabert B, Geczik AM, Bauer DC, Buist DSM, Cauley JA, Falk RT, Gierach GL, Hue TF, Lacey JV, LaCroix AZ, Michels KA, Tice JA, Xu X, Brinton LA, Dallal CM. Association of Endogenous Pregnenolone, Progesterone, and Related Metabolites with Risk of Endometrial and Ovarian Cancers in Postmenopausal Women: The B ∼FIT Cohort. Cancer Epidemiol Biomarkers Prev 2021; 30:2030-2037. [PMID: 34465588 DOI: 10.1158/1055-9965.epi-21-0669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/19/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Postmenopausal pregnenolone and/or progesterone levels in relation to endometrial and ovarian cancer risks have been infrequently evaluated. To address this, we utilized a sensitive and reliable assay to quantify prediagnostic levels of seven markers related to endogenous hormone metabolism. METHODS Hormones were quantified in baseline serum collected from postmenopausal women in a cohort study nested within the Breast and Bone Follow-up to the Fracture Intervention Trial (B∼FIT). Women using exogenous hormones at baseline (1992-1993) were excluded. Incident endometrial (n = 65) and ovarian (n = 67) cancers were diagnosed during 12 follow-up years and compared with a subcohort of 345 women (no hysterectomy) and 413 women (no oophorectomy), respectively. Cox models with robust variance were used to estimate cancer risk. RESULTS Circulating progesterone levels were not associated with endometrial [tertile (T)3 vs. T1 HR (95% confidence interval): 1.87 (0.85-4.11); P trend = 0.17] or ovarian cancer risk [1.16 (0.58-2.33); 0.73]. Increasing levels of the progesterone-to-estradiol ratio were inversely associated with endometrial cancer risk [T3 vs. T1: 0.29 (0.09-0.95); 0.03]. Increasing levels of 17-hydroxypregnenolone were inversely associated with endometrial cancer risk [0.40 (0.18-0.91); 0.03] and positively associated with ovarian cancer risk [3.11 (1.39-6.93); 0.01]. CONCLUSIONS Using sensitive and reliable assays, this study provides novel data that endogenous progesterone levels are not strongly associated with incident endometrial or ovarian cancer risks. 17-hydroxypregnenolone was positively associated with ovarian cancer and inversely associated with endometrial cancer. IMPACT While our results require replication in large studies, they provide further support of the hormonal etiology of endometrial and ovarian cancers.
Collapse
Affiliation(s)
- Britton Trabert
- Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland. .,Department of Obstetrics and Gynecology, University of Utah, and Cancer Control and Population Sciences Research Program, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Ashley M Geczik
- Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland
| | - Doug C Bauer
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California.,Department of Medicine, University of California San Francisco, San Francisco, California
| | - Diana S M Buist
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington
| | - Jane A Cauley
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Roni T Falk
- Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland
| | | | - Trisha F Hue
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California
| | - James V Lacey
- Division of Health Analytics, Department of Computational and Quantitative Medicine, City of Hope, Duarte, California
| | - Andrea Z LaCroix
- Division of Epidemiology, Department of Family and Preventive Medicine, University of California San Diego, San Diego, California
| | - Kara A Michels
- Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland
| | - Jeffrey A Tice
- Department of Medicine, University of California San Francisco, San Francisco, California
| | - Xia Xu
- Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Louise A Brinton
- Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland
| | - Cher M Dallal
- School of Public Health, University of Maryland, College Park, Maryland
| |
Collapse
|
15
|
Effect of Interaction between 17β-Estradiol, 2-Methoxyestradiol and 16α-Hydroxyestrone with Chromium (VI) on Ovary Cancer Line SKOV-3: Preliminary Study. Molecules 2020; 25:molecules25215214. [PMID: 33182506 PMCID: PMC7665134 DOI: 10.3390/molecules25215214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/22/2020] [Accepted: 11/06/2020] [Indexed: 01/06/2023] Open
Abstract
Ovarian cancer is the leading cause of death from gynecologic malignancies. Some estrogens, as well as xenoestrogens, such as chromium (VI) (Cr(VI)), are indicated as important pathogenic agents. The objective of this study was to evaluate the role of estradiol and some its metabolites upon exposure to the metalloestrogen Cr(VI) in an in vitro model. The changes in cell viability of malignant ovarian cancer cells (SKOV-3 resistant to cisplatin) exposed to 17β-estradiol (E2) and its two metabolites, 2-methoxyestradiol (2-MeOE2) and 16α-hydroxyestrone (16α-OHE1), upon exposure to potassium chromate (VI) and its interactions were examined. The single and mixed models of action, during short and long times of incubation with estrogens, were applied. The different effects (synergism and antagonism) of estrogens on cell viability in the presence of Cr(VI) was observed. E2 and 16α-OHE1 caused a synergistic effect after exposure to Cr(VI). 2-MeOE2 showed an antagonistic effect on Cr(VI). The examined estrogens could be ranked according to the most protective effect or least toxicity in the order: 2-MeOE2 > E2 > 16α-OHE1. Early pre-incubation (24 h or 7 days) of cells with estrogens caused mostly an antagonistic effect-protective against the toxic action of Cr(VI). The beneficial action of estrogens on the toxic effect of Cr(VI), in the context of the risk of ovarian cancer, seems to be important and further studies are needed.
Collapse
|
16
|
Davis SR, Martinez-Garcia A, Robinson PJ, Handelsman DJ, Desai R, Wolfe R, Bell RJ. Estrone Is a Strong Predictor of Circulating Estradiol in Women Age 70 Years and Older. J Clin Endocrinol Metab 2020; 105:5866468. [PMID: 32614391 PMCID: PMC7394338 DOI: 10.1210/clinem/dgaa429] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/29/2020] [Indexed: 11/19/2022]
Abstract
IMPORTANCE After menopause, estradiol (E2) is predominately an intracrine hormone circulating in very low serum concentrations. OBJECTIVE The objective of this work is to examine determinants of E2 concentrations in women beyond age 70 years. DESIGN AND SETTING A cross-sectional, community-based study was conducted. PARTICIPANTS A total of 5325 women participated, with a mean age of 75.1 years (± 4.2 years) and not using any sex steroid, antiandrogen/estrogen, glucocorticoid, or antiglycemic therapy. MAIN OUTCOME MEASURES Sex steroids were measured by liquid chromatography-tandem mass spectrometry. Values below the limit of detection (LOD; E2 11 pmol/L [3 pg/mL] were assigned a value of LOD/√2 to estimate total E2. RESULTS E2 and estrone (E1) were below the LOD in 66.1% and 0.9% of women, respectively. The median (interdecile ranges) for E1 and detectable E2 were 181.2 pmol/L (range, 88.7-347.6 pmol/L) and 22.0 pmol/L (range, 11.0-58.7 pmol/L). Women with undetectable E2 vs detectable E2 were older (median age 74.1 years vs 73.8, P = .02), leaner (median body mass index [BMI] 26.8 kg/m2 vs 28.5, P < .001), and had lower E1, testosterone and DHEA concentrations (P < .001). A linear regression model, including age, BMI, E1, and testosterone, explained 20.9% of the variation in total E2, but explained only an additional 1.2% of variation over E1 alone. E1 and testosterone made significant contributions (r2 = 0.162, P < .001) in a model for the subset of women with detectable E2. CONCLUSIONS Our findings support E1 as a principal circulating estrogen and demonstrate a robust association between E1 and E2 concentrations in postmenopausal women. Taken together with prior evidence for associations between E1 and health outcomes, E1 should be included in studies examining associations between estrogen levels and health outcomes in postmenopausal women.
Collapse
Affiliation(s)
- Susan R Davis
- Women’s Health Research Program, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Correspondence and Reprint Requests: Susan Davis, MD, PhD, Women’s Health Research Program, School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne 3004, Victoria, Australia. E-mail:
| | - Alejandra Martinez-Garcia
- Women’s Health Research Program, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Endocrinology, Division of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Penelope J Robinson
- Women’s Health Research Program, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - David J Handelsman
- ANZAC Research Institute, University of Sydney, New South Wales, Australia
| | - Reena Desai
- ANZAC Research Institute, University of Sydney, New South Wales, Australia
| | - Rory Wolfe
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Robin J Bell
- Women’s Health Research Program, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | | |
Collapse
|
17
|
Khan WA, Alsamghan AS, Khan MWA. Endometrial cancer patients have high affinity antibodies for estrogen metabolite-receptor aggregate: A potential biomarker for EC. J Obstet Gynaecol Res 2020; 46:2115-2125. [PMID: 32761720 DOI: 10.1111/jog.14413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/26/2020] [Accepted: 07/10/2020] [Indexed: 12/29/2022]
Abstract
AIM Elevated levels of 16α-hydroxyestrone (16α-OHE1 ) have been described in endometrial cancer (EC) and estrogen receptors (ER) expressed in endometrial tissue, but research on their combined role is lacking. We aimed to investigate the affinity and binding specificity of EC antibodies against the 16α-OHE1 -ERα aggregate in the serum of EC patients. Specificities of EC antibodies were also evaluated according to various clinical characteristics found in these cancer patients. METHODS The binding specificity and affinity of EC antibodies against 16α-OHE1 -ERα in the serum of 120 EC patients were evaluated by direct binding and competition ELISA and quantitative precipitation titration. Binding of EC antibodies was also determined according to various clinical characteristics in EC patients through competition ELISA. RESULTS Antibodies from EC patients demonstrated high recognition of 16α-OHE1 -ERα compared to ERα (P < 0.05) or 16α-OHE1 (P < 0.001). The relative affinity of EC IgG was 1.49 × 10-7 M, 1.34 × 10-6 M and 1.13 × 10-6 M for 16α-OHE1 -ERα, ERα and 16α-OHE1 , respectively. Several factors, such as obesity, postmenopausal status, use of hormonal therapy, ER and progesterone receptor (PR) status, low 2-OHE1 /16α-OHE1 ratio, chemotherapy and hypertension, augment the production of antibodies against 16α-OHE1 -ERα in EC patients. CONCLUSION 16α-OHE1 -ERα is a high-affinity antigen for EC antibodies in the serum of EC patients and might function as a biomarker for this disease. Furthermore, several factors enhanced the production of antibodies against 16α-OHE1 -ERα in the sera of these EC patients.
Collapse
Affiliation(s)
- Wahid Ali Khan
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, KSA
| | - Awad Saeed Alsamghan
- Department of Family and Community Medicine, College of Medicine, King Khalid University, Abha, KSA
| | - Mohd Wajid Ali Khan
- Department of Clinical Laboratory Science, College of Applied Medical Science, University of Hail, Hail, KSA.,Molecular Diagnostic and Personalised Therapeutics Unit, University of Hail, Hail, KSA
| |
Collapse
|
18
|
Wang Y, Chen R, Gan Y, Ying S. The roles of PAD2- and PAD4-mediated protein citrullination catalysis in cancers. Int J Cancer 2020; 148:267-276. [PMID: 33459350 DOI: 10.1002/ijc.33205] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
Abstract
Peptidylarginine deiminases (PADs) catalyze the conversion of arginine residues to citrulline residues on target proteins in the presence of calcium ions. This elaborate type of posttranslational modification is termed citrullination. PADs may regulate gene transcriptional activity via histone citrullination. There has been an increasing appreciation for the roles of PADs in a wide variety of biological processes. In this review article, we summarize recent evidence indicating that PADs and citrullinated proteins are involved in several physiological and pathological processes related to cancer. Of particular interest is that PAD2 and PAD4 exhibit characteristic expression levels, activities and specific biological effects in diverse types of cancer. We also list several PAD inhibitors, propose the possible mechanisms underlying the biological actions of PAD-mediated protein citrullination in experimental models and discuss the potential therapeutic value of PADs and their inhibitors for disease diagnosis and treatment.
Collapse
Affiliation(s)
- Yanbin Wang
- Hangzhou Medical College, Hangzhou, Zhejiang, China.,Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, China
| | - Riping Chen
- Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yihan Gan
- Hangzhou Medical College, Hangzhou, Zhejiang, China.,Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, China
| | - Shibo Ying
- Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
19
|
Li S, Chen Y, Xie L, Meng Y, Zhu L, Chu H, Gu D, Zhang Z, Du M, Wang M. Sex hormones and genetic variants in hormone metabolic pathways associated with the risk of colorectal cancer. ENVIRONMENT INTERNATIONAL 2020; 137:105543. [PMID: 32059146 DOI: 10.1016/j.envint.2020.105543] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/02/2020] [Accepted: 02/02/2020] [Indexed: 06/10/2023]
Abstract
OBJECTIVE The different incidence of colorectal cancer between the sexes suggests that sex hormones may be involved in the susceptibility to colorectal cancer. The association between sex hormones and genetic variants in hormone metabolic pathways and the colorectal cancer risk remains unclear. METHODS We detected sex hormone levels in plasma from colorectal cancer patients and controls in males by ultra-high-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). We evaluated the clinical significance of sex hormones on colorectal cancer diagnosis with the area under the receiver operating characteristic curve (AUC). The role of genetic variants in hormone metabolic pathways in the colorectal cancer risk was assessed by a logistic regression model. The biological functions were detected by luciferase reporter assays and cell behavior experiments. RESULTS We found that 2-methoxyestrone (2-MeO-E1) was highly expressed in cases (PFDR = 3.48 × 10-19). The expression of 2-MeO-E1 in plasma showed improved accuracy for predicting colorectal cancer (AUC = 0.88). In the 2-MeO-E1 metabolic pathway, rs165599 in COMT was significantly associated with an increased risk of colorectal cancer (P = 0.009). Mechanistically, we found that the rs165599 G allele could decrease the binding ability of miR-22-3p to the COMT 3'-UTR. Furthermore, knockdown of COMT inhibited cell proliferation, induced cell apoptosis and arrested the cell cycle in the G1 phase. CONCLUSION This is the first study to show that 2-MeO-E1 and a genetic variant in COMT contribute to the susceptibility to colorectal cancer. These results shed light on the different incidence of colorectal cancer between the sexes.
Collapse
Affiliation(s)
- Shuwei Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yehua Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lisheng Xie
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Infection Control, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai, China
| | - Yixuan Meng
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lingjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haiyan Chu
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Dongying Gu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zhengdong Zhang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Mulong Du
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Meilin Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
20
|
Trabert B, Coburn SB, Falk RT, Manson JE, Brinton LA, Gass ML, Kuller LH, Rohan TE, Pfeiffer RM, Qi L, Stefanick ML, Wentzensen N, Anderson GL, Xu X. Circulating estrogens and postmenopausal ovarian and endometrial cancer risk among current hormone users in the Women's Health Initiative Observational Study. Cancer Causes Control 2019; 30:1201-1211. [PMID: 31542834 PMCID: PMC6785392 DOI: 10.1007/s10552-019-01233-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/11/2019] [Indexed: 11/28/2022]
Abstract
PURPOSE Menopausal hormone therapy (MHT) use induces alterations in circulating estrogens/estrogen metabolites, which may contribute to the altered risk of reproductive tract cancers among current users. Thus, the current study assessed associations between circulating estrogens/estrogen metabolites and ovarian and endometrial cancer risk among MHT users. METHODS We conducted a nested case-control study among postmenopausal women using MHT at baseline in the Women's Health Initiative Observational Study (179 ovarian cancers, 396 controls; 230 endometrial cancers, 253 controls). Multivariable logistic regression was utilized to estimate odds ratios and 95% confidence intervals overall and by subtype. RESULTS Estrogen/estrogen metabolite levels were not associated with overall or serous ovarian cancer risk, examined separately. However, unconjugated estradiol was positively associated with non-serous ovarian cancer risk [quintile 5 vs. quintile 1: 3.01 (1.17-7.73); p-trend = 0.03; p-het < 0.01]. Endometrial cancer risk was unrelated to estrogen/estrogen metabolite levels among women who took combined estrogen/progestin therapy (EPT). CONCLUSIONS These findings provide novel evidence that may support a heterogeneous hormonal etiology across ovarian cancer subtypes. Circulating estrogens did not influence endometrial cancer risk among women with EPT-induced high-estrogen levels. Larger studies are needed to delineate the relationship between ovarian/endometrial cancer subtypes and estrogen levels in the context of MHT use.
Collapse
Affiliation(s)
- Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892-9768, USA.
| | - Sally B Coburn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892-9768, USA
| | - Roni T Falk
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892-9768, USA
| | - JoAnn E Manson
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Louise A Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892-9768, USA
| | - Margery L Gass
- Women's Health Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lewis H Kuller
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thomas E Rohan
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892-9768, USA
| | - Lihong Qi
- Public Health Sciences, School of Medicine, UC Davis, Sacramento, CA, USA
| | - Marcia L Stefanick
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892-9768, USA
| | - Garnet L Anderson
- Division of Public Health Sciences, Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Xia Xu
- Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick, MD, USA
| |
Collapse
|
21
|
Anh NH, Long NP, Kim SJ, Min JE, Yoon SJ, Kim HM, Yang E, Hwang ES, Park JH, Hong SS, Kwon SW. Steroidomics for the Prevention, Assessment, and Management of Cancers: A Systematic Review and Functional Analysis. Metabolites 2019; 9:E199. [PMID: 31546652 PMCID: PMC6835899 DOI: 10.3390/metabo9100199] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/09/2019] [Accepted: 09/17/2019] [Indexed: 02/07/2023] Open
Abstract
Steroidomics, an analytical technique for steroid biomarker mining, has received much attention in recent years. This systematic review and functional analysis, following the PRISMA statement, aims to provide a comprehensive review and an appraisal of the developments and fundamental issues in steroid high-throughput analysis, with a focus on cancer research. We also discuss potential pitfalls and proposed recommendations for steroidomics-based clinical research. Forty-five studies met our inclusion criteria, with a focus on 12 types of cancer. Most studies focused on cancer risk prediction, followed by diagnosis, prognosis, and therapy monitoring. Prostate cancer was the most frequently studied cancer. Estradiol, dehydroepiandrosterone, and cortisol were mostly reported and altered in at least four types of cancer. Estrogen and estrogen metabolites were highly reported to associate with women-related cancers. Pathway enrichment analysis revealed that steroidogenesis; androgen and estrogen metabolism; and androstenedione metabolism were significantly altered in cancers. Our findings indicated that estradiol, dehydroepiandrosterone, cortisol, and estrogen metabolites, among others, could be considered oncosteroids. Despite noble achievements, significant shortcomings among the investigated studies were small sample sizes, cross-sectional designs, potential confounding factors, and problematic statistical approaches. More efforts are required to establish standardized procedures regarding study design, analytical procedures, and statistical inference.
Collapse
Affiliation(s)
- Nguyen Hoang Anh
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | | | - Sun Jo Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Jung Eun Min
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Sang Jun Yoon
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Hyung Min Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Eugine Yang
- College of Pharmacy, Ewha Womans University, Seoul 03760, Korea.
| | - Eun Sook Hwang
- College of Pharmacy, Ewha Womans University, Seoul 03760, Korea.
| | - Jeong Hill Park
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Korea.
| | - Sung Won Kwon
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
22
|
Denver N, Khan S, Homer NZM, MacLean MR, Andrew R. Current strategies for quantification of estrogens in clinical research. J Steroid Biochem Mol Biol 2019; 192:105373. [PMID: 31112747 PMCID: PMC6726893 DOI: 10.1016/j.jsbmb.2019.04.022] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/24/2019] [Accepted: 04/29/2019] [Indexed: 12/22/2022]
Abstract
Estrogens and their bioactive metabolites play key roles in regulating diverse processes in health and disease. In particular, estrogens and estrogenic metabolites have shown both protective and non-protective effects on disease pathobiology, implicating the importance of this steroid pathway in disease diagnostics and monitoring. All estrogens circulate in a wide range of concentrations, which in some patient cohorts can be extremely low. However, elevated levels of estradiol are reported in disease. For example, in pulmonary arterial hypertension (PAH) elevated levels have been reported in men and postmenopausal women. Conventional immunoassay techniques have come under scrutiny, with their selectivity, accuracy and precision coming into question. Analytical methodologies such as gas and liquid chromatography coupled to single and tandem mass spectrometric approaches (GC-MS, GC-MS/MS, LC-MS and LC-MS/MS) have been developed to quantify endogenous estrogens and in some cases their bioactive metabolites in biological fluids such as urine, serum, plasma and saliva. Liquid-liquid or solid-phase extraction approaches are favoured with derivatization remaining a necessity for detection in lower volumes of sample. The limits of quantitation of individual assays vary but are commonly in the range of 0.5-5 pg/mL for estrone and estradiol, with limits for their bioactive metabolites being higher. This review provides an overview of current approaches for measurement of unconjugated estrogens in biological matrices by MS, highlighting the advances in this field and the challenges remaining for routine use in the clinical and research environment.
Collapse
Affiliation(s)
- Nina Denver
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, University Avenue, Glasgow, G12 8QQ, United Kingdom; Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, United Kingdom.
| | - Shazia Khan
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom; University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh, UK, EH16 4TJ.
| | - Natalie Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom.
| | - Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, United Kingdom.
| | - Ruth Andrew
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom; University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47, Little France Crescent, Edinburgh, UK, EH16 4TJ.
| |
Collapse
|
23
|
Yang B, Chen R, Liang X, Shi J, Wu X, Zhang Z, Chen X. Estrogen Enhances Endometrial Cancer Cells Proliferation by Upregulation of Prohibitin. J Cancer 2019; 10:1616-1621. [PMID: 31205517 PMCID: PMC6548001 DOI: 10.7150/jca.28218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 01/13/2019] [Indexed: 12/12/2022] Open
Abstract
Estrogen plays an essential role in type I endometrial cancer cell proliferation. Despite great progresses in the etiology has been obtained in the past, however, the molecular mechanisms remain to be fully clarified. Prohibitin has been demonstrated involvement in multiple cancers' development. If it also contributes to estrogen-driven endometrial cancer proliferation is not clear. IHC assay result display that prohibitin overexpressed in endometrial cancer tissue and associated with the poor prognosis; Western blot assay detect that upregulated prohibitin expression with dose- and time-dependent manners. The cellular growth was monitored with SRB assay which demonstrate that knockdown prohibitin attenuated estrogen-induced proliferation. Ubiquitination assay finds estrogen increased prohibitin level through stabilizing prohibitin protein via inhibition of ubiquitination, while estrogen-induced protein expression was mediated by estrogen receptor. Our findings provide a new insight on the mechanism of estrogen-induced proliferation, implying the possibility of using prohibitin as a potential therapeutic target for the treatment of endometrial cancer.
Collapse
Affiliation(s)
- Bin Yang
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital, Baoshan Branch, Shanghai, 201900, China.,Jiangxi Medical College, Nanchang University, Nanchang, 330000, China
| | - Ruiying Chen
- Department of Cervical Diseases, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Xiaoyan Liang
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital, Baoshan Branch, Shanghai, 201900, China
| | - Jiayan Shi
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital, Baoshan Branch, Shanghai, 201900, China
| | - Xiaomei Wu
- Reproductive Medicine, Department of Obstetrics and Gynecology,Shanghai First people's Hospital, Shanghai Jiaotong University, Shanghai, 201600,China.,Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, 201600, China
| | - Zhenbo Zhang
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital, Baoshan Branch, Shanghai, 201900, China.,Jiangxi Medical College, Nanchang University, Nanchang, 330000, China.,Reproductive Medicine, Department of Obstetrics and Gynecology,Shanghai First people's Hospital, Shanghai Jiaotong University, Shanghai, 201600,China.,Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, 201600, China
| | - Xiong Chen
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital, Baoshan Branch, Shanghai, 201900, China.,Jiangxi Medical College, Nanchang University, Nanchang, 330000, China
| |
Collapse
|
24
|
Han L, Du J, Zhao L, Sun C, Wang Q, Tuo X, Hou H, Liu Y, Wang Q, Ulain Q, Lv S, Zhang G, Song Q, Li Q. An Efficacious Endometrial Sampler for Screening Endometrial Cancer. Front Oncol 2019; 9:67. [PMID: 30838173 PMCID: PMC6389657 DOI: 10.3389/fonc.2019.00067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 01/24/2019] [Indexed: 11/25/2022] Open
Abstract
Recently, the research on early detection of precancerous change and endometrial carcinoma has been focusing on minimally invasive procedures for screening. On this basis, we aim to verify the feasibility of endometrial samplers for screening endometrial cancer using Li Brush. We recruited patients undergoing hysterectomy for different diseases from the Inpatient Department of the Department of Obstetrics and Gynecology. Before surgery, endometrial cells were collected by Li Brush. The cytopathologic diagnosis from Li Brush and the histopathologic diagnosis from hysterectomy in the same patient were compared to calculate sensitivity (Se), specificity (Sp), false-negative rate (FNR), false-positive rate (FPR), positive predictive value (PV+) %, and negative predictive value (PV-). The research enrolled 293 women into this self-controlled trial. According to the hypothesis test of paired four lattices, we obtained the following indicators: Se 92.73, Sp 98.15, FNR 7.27, FPR 1.85, PV+92.73, and PV-98.15%. The endometrial sampler Li Brush is an efficacious instrument for screening endometrial cancer.
Collapse
Affiliation(s)
- Lu Han
- Center for Single-Cell Biology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jiang Du
- Department of Gynecology and Obstetrics, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Lanbo Zhao
- Guipei 77, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Chao Sun
- Center for Single-Cell Biology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qi Wang
- Center for Single-Cell Biology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoqian Tuo
- Center for Single-Cell Biology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Huilian Hou
- Department of Pathology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yu Liu
- Department of Pathology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qing Wang
- Center for Single-Cell Biology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qurat Ulain
- Center for Single-Cell Biology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Shulan Lv
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Guanjun Zhang
- Department of Pathology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qing Song
- Center for Single-Cell Biology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, GA, United States
| | - Qiling Li
- Center for Single-Cell Biology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
25
|
An D, Song Z, Yi Y, Zhang Q, Liu J, Zhang Y, Zhou J, Zhao G, Cong D, Li N, Lu Y, Chen X, Zhao D. Oroxylin A, a methylated metabolite of baicalein, exhibits a stronger inhibitory effect than baicalein on the CYP1B1-mediated carcinogenic estradiol metabolite formation. Phytother Res 2019; 33:1033-1043. [PMID: 30680817 DOI: 10.1002/ptr.6297] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 12/16/2022]
Abstract
Human cytochrome P450 1B1 (CYP1B1)-mediated formation of 4-hydroxyestradiol (4-OHE2) from 17β-estradiol plays an important role in the progression of human breast cancer, while the biotransformation of 17β-estradiol to 2-hydroxyestradiol mediated by cytochrome P450 1A1 (CYP1A1) is considered as a less harmful pathway. In this study, inhibitory effects of flavonoids baicalein and oroxylin A, a metabolite of baicalein in human body, on CYP1A1 and 1B1 activities were investigated in vitro. The inhibition intensities of baicalein and oroxylin A towards CYP1B1 were greater than towards CYP1A1 with a mixed mechanism. In addition, oroxylin A showed a stronger inhibitory effect than baicalein towards the CYP1B1-mediated 17β-estradiol 4-hydroxylation, with the IC50 values of 0.0146 and 2.27 μM, respectively. Docking studies elucidated that oroxylin A had a stronger binding affinity than baicalein for CYP1B1. In MCF-7 cells, compared with baicalein-treated groups, oroxylin A with lower doses decreased and increased the formation of 4-OHE2 and 2-hydroxyestradiol, respectively, with a preferential induction of mRNA of CYP1A1 over CYP1B1. In conclusion, this study demonstrated that oroxylin A showed a stronger inhibitory effect than baicalein on CYP1B1-mediated 4-OHE2 formation in MCF-7 cells, providing crucial implications for their possibly preventive/therapeutic potential against breast cancer via inhibition of CYP1B1, particularly of oroxylin A.
Collapse
Affiliation(s)
- Dongchen An
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Zhongjin Song
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Yingyue Yi
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Qing Zhang
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Jinfeng Liu
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Yongjie Zhang
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Jing Zhou
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Guanghui Zhao
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Danhua Cong
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Ning Li
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Yang Lu
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Xijing Chen
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Di Zhao
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
26
|
Audet-Delage Y, Grégoire J, Caron P, Turcotte V, Plante M, Ayotte P, Simonyan D, Villeneuve L, Guillemette C. Estradiol metabolites as biomarkers of endometrial cancer prognosis after surgery. J Steroid Biochem Mol Biol 2018; 178:45-54. [PMID: 29092787 DOI: 10.1016/j.jsbmb.2017.10.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/23/2017] [Accepted: 10/26/2017] [Indexed: 12/18/2022]
Abstract
Endometrial cancer (EC) is the most common gynecologic malignancy prevailing after menopause. Defining steroid profiles may help predict the risk of recurrence after hysterectomy, which remains limited due to the lack of reliable markers. Adrenal precursors, androgens, parent estrogens and catechol estrogen metabolites were measured by mass spectrometry (MS) in preoperative serums and those collected one month after hysterectomy from 246 newly diagnosed postmenopausal EC cases. We also examined the associations between steroid hormones and EC status by including 110 healthy postmenopausal women. Steroid concentrations were analyzed in relation to clinicopathological features, recurrence and overall survival (OS). The mean follow-up time was 65.5 months and 26 patients experienced relapse after surgery for a recurrence incidence of 10.6% (6.4% Type I and 29.5% Type II). Recurrence and OS were related to a more aggressive disease but not linked to body mass index. Preoperative levels of estriol (E3) and estrone-sulfate (E1-S) were inversely associated with recurrence in a multivariate logistic regression analysis (Hazard ratios (HRs) of 0.31, P=0.039 and 3.01, P=0.024; respectively). All circulating steroids declined considerably after surgery almost reaching those of healthy women, except 4-methoxy-E2 (4MeO-E2) for which postoperative levels increased by 35% and were associated to a 68% decreased risk of recurrence (HR=0.32, P=0.015). Women diagnosed with both histological types of EC present significantly higher levels of steroids, in support of their mitogenic effects. The estrogen precursor E1-S, the anticancer metabolite 4MeO-E2, and E3 that exert mixed antagonist and agonist estrogenic activities and immunological effects, are potential independent prognostic factors.
Collapse
Affiliation(s)
- Yannick Audet-Delage
- Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center and Faculty of Pharmacy, Laval University, Québec, Canada
| | - Jean Grégoire
- Gynecologic Oncology Service, CHU de Québec, and Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Laval University, Québec, Canada
| | - Patrick Caron
- Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center and Faculty of Pharmacy, Laval University, Québec, Canada
| | - Véronique Turcotte
- Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center and Faculty of Pharmacy, Laval University, Québec, Canada
| | - Marie Plante
- Gynecologic Oncology Service, CHU de Québec, and Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Laval University, Québec, Canada
| | - Pierre Ayotte
- CHU de Québec Research Center, and Department of Social and Preventive Medicine, Faculty of Medicine, Laval University, Québec, Canada
| | - David Simonyan
- Statistical and Clinical Research Platform, CHU de Québec Research Center, Québec, Canada
| | - Lyne Villeneuve
- Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center and Faculty of Pharmacy, Laval University, Québec, Canada
| | - Chantal Guillemette
- Centre Hospitalier Universitaire de Québec (CHU de Québec) Research Center and Faculty of Pharmacy, Laval University, Québec, Canada; Canada Research Chair in Pharmacogenomics, Canada.
| |
Collapse
|
27
|
Yeganeh L, Harrison C, Vincent AJ, Teede H, Boyle JA. Effects of lifestyle modification on cancer recurrence, overall survival and quality of life in gynaecological cancer survivors: A systematic review and meta-analysis. Maturitas 2018; 111:82-89. [PMID: 29673836 DOI: 10.1016/j.maturitas.2018.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 03/12/2018] [Indexed: 01/24/2023]
Abstract
The benefits of lifestyle interventions for women who have survived gynaecological cancer (GC) remain unclear. This systematic review aimed to determine the effect of lifestyle interventions on cancer recurrence, overall survival and quality of life (QoL) in women with GC. We searched Medline, Embase, PsycINFO and EBM Reviews from June to July 2016 to identify relevant literature. We included randomized controlled trials in which a lifestyle intervention (diet, weight loss, physical activity and/or behavioural interventions) were compared with a control condition (usual care, placebo or other lifestyle interventions) in women who had survived endometrial or ovarian cancer. Primary outcomes included cancer recurrence and overall survival and the secondary outcome was QoL. Data extraction and risk-of-bias assessment were performed by two independent reviewers. A random-effects meta-analysis model was used to calculate mean differences (md) and 95% confidence intervals (CI). The literature search yielded 928 citations and three trials met the inclusion criteria. No randomized controlled trial assessed the effect of lifestyle interventions on cancer recurrence or survival. Meta-analysis of two randomized controlled trials on the effect of lifestyle interventions on total QoL at 3 or 6 months post-intervention showed no significant difference between intervention and control groups [(md; 1.60; 95% CI, -1.65 to 4.85) and (md; 2.07; 95% CI, -1.80 to 5.94), respectively]. That is, lifestyle intervention had no effect on overall QoL or individual QoL domains (physical, emotional, social wellbeing and fatigue) in GC survivors. Systematic review registration: PROSPERO CRD42016043719.
Collapse
Affiliation(s)
- Ladan Yeganeh
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia.
| | - Cheryce Harrison
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia.
| | - Amanda J Vincent
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia; Menopause Unit, Monash Health, Melbourne, Victoria, Australia.
| | - Helena Teede
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia; Diabetes and Vascular Medicine Unit, Monash Health, Melbourne, Victoria, Australia; Monash Partners Academic Health Sciences Centre, Melbourne, Victoria, Australia.
| | - Jacqueline A Boyle
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia; Menopause Unit, Monash Health, Melbourne, Victoria, Australia.
| |
Collapse
|
28
|
|
29
|
Chen S, Chen Y, Ma S, Zheng R, Zhao P, Zhang L, Liu Y, Yu Q, Deng Q, Zhang K. Dietary fibre intake and risk of breast cancer: A systematic review and meta-analysis of epidemiological studies. Oncotarget 2018; 7:80980-80989. [PMID: 27829237 PMCID: PMC5348370 DOI: 10.18632/oncotarget.13140] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/27/2016] [Indexed: 12/13/2022] Open
Abstract
Current evidence from randomised controlled trials on the effects of dietary fibre intake on breast cancer risk is inconsistent. We conducted a meta-analysis to determine the effectiveness of dietary fibre intake in reducing breast cancer risk. We searched for prospective and case-control studies on dietary fibre intake and breast cancer risk in the English language through March 2016. Twenty-four epidemiologic studies obtained through the PubMed, Embase, Web of Science, and Cochrane Library databases were systematically reviewed. A random-effects model was used to compute the pooled risk estimates by extracting the risk estimate of the highest and lowest reported categories of intake from each study. The meta-analyses showed a 12% decrease in breast cancer risk with dietary fibre intake. The association between dietary fibre intake and breast cancer risk was significant when stratified according to Jadad scores, study types, and menopause status. Dose-response analysis showed that every 10 g/d increment in dietary fibre intake was associated with a 4% reduction in breast cancer risk, and little evidence of publication bias was found. Thus, dietary fibre consumption is significantly associated with a reduced risk of breast cancer, particularly in postmenopausal women.
Collapse
Affiliation(s)
- Sumei Chen
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China
| | - Yuanyuan Chen
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China
| | - Shenglin Ma
- Affiliated Hangzhou First People's Hospital, Hangzhou, Zhejiang 310006, China.,Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou, Zhejiang 310006, China
| | - Ruzhen Zheng
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China
| | - Pengjun Zhao
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China
| | - Lidan Zhang
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China
| | - Yuehua Liu
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China
| | - Qingqing Yu
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China
| | - Qinghua Deng
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China.,Affiliated Hangzhou First People's Hospital, Hangzhou, Zhejiang 310006, China
| | - Ke Zhang
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China
| |
Collapse
|
30
|
Audet-Delage Y, Villeneuve L, Grégoire J, Plante M, Guillemette C. Identification of Metabolomic Biomarkers for Endometrial Cancer and Its Recurrence after Surgery in Postmenopausal Women. Front Endocrinol (Lausanne) 2018; 9:87. [PMID: 29593653 PMCID: PMC5857535 DOI: 10.3389/fendo.2018.00087] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/23/2018] [Indexed: 11/24/2022] Open
Abstract
Endometrial cancer (EC) is the most frequent gynecological cancer in developed countries. Most EC occurs after menopause and is diagnosed as endometrioid (type I) carcinomas, which exhibit a favorable prognosis. In contrast, non-endometrioid (type II) carcinomas such as serous tumors have a poor prognosis. Our goal was to identify novel blood-based markers associated with EC subtypes and recurrence after surgery in postmenopausal women. Using mass spectrometry-based untargeted metabolomics, we examined preoperative serum metabolites among control women (n = 18) and those with non-recurrent (NR) and recurrent (R) cases of type I endometrioid (n = 24) and type II serous (n = 12) carcinomas. R and NR cases were similar with respect to pathological characteristics, body mass index, and age. A total of 1,592 compounds were analyzed including 14 different lipid classes. When we compared EC cases with controls, 137 metabolites were significantly different. A combination of spermine and isovalerate resulted in an age-adjusted area under the receiver-operating characteristic curve (AUCadj) of 0.914 (P < 0.001) for EC detection. The combination of 2-oleoylglycerol and TAG42:2-FA12:0 allowed the distinction of R cases from NR cases with an AUCadj of 0.901 (P < 0.001). Type I R cases were also characterized by much lower levels of bile acids and elevated concentrations of phosphorylated fibrinogen cleavage peptide, whereas type II R cases displayed higher levels of ceramides. The findings from our pilot study provide a detailed metabolomics study of EC and identify putative serum biomarkers for defining clinically relevant risk groups.
Collapse
Affiliation(s)
- Yannick Audet-Delage
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Lyne Villeneuve
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Jean Grégoire
- Gynecologic Oncology Service, CHU de Québec, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Marie Plante
- Gynecologic Oncology Service, CHU de Québec, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Chantal Guillemette
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Pharmacy, Laval University, Québec, QC, Canada
- Canada Research Chair in Pharmacogenomi, Laval University, Québec, QC, Canada
- *Correspondence: Chantal Guillemette,
| |
Collapse
|
31
|
Playdon MC, Coburn SB, Moore SC, Brinton LA, Wentzensen N, Anderson G, Wallace R, Falk RT, Pfeiffer R, Xu X, Trabert B. Alcohol and oestrogen metabolites in postmenopausal women in the Women's Health Initiative Observational Study. Br J Cancer 2017; 118:448-457. [PMID: 29235567 PMCID: PMC5808032 DOI: 10.1038/bjc.2017.419] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/25/2017] [Accepted: 10/26/2017] [Indexed: 12/31/2022] Open
Abstract
Background: Alcohol consumption is associated with an increased risk of several cancers. Potential mechanisms include altered oestrogen metabolism. Parent oestrogens metabolise into alternate pathways of oestrogen metabolites that may have variable effects on cancer pathogenesis. We examined associations of alcohol consumption with circulating oestrogen/oestrogen metabolites in postmenopausal women in the Women’s Health Initiative (WHI)-Observational Study (OS). Methods: We conducted a cross-sectional analysis of prediagnosis ovarian/endometrial cancer case-control data within WHI-OS (N=1864). Alcohol consumption was measured by validated food frequency questionnaire. Fasting serum parent oestrogens/oestrogen metabolites were assayed using liquid chromatography tandem mass-spectrometry. Geometric mean analyte concentrations (GM, pmol l−1) were calculated by alcohol category using inverse-probability weighted linear regression, adjusting for venepuncture age/year, race, smoking, body mass index, years since menopause, oral contraceptive duration, caffeine intake, and physical activity. Results: There was evidence for a positive association between alcohol consumption and oestrone, oestradiol and 2-hydroxylation oestrogen metabolite concentrations among menopausal hormone therapy (MHT) users. We observed an association between liquor consumption and parent oestrogens among non-MHT users, who consumed larger doses of liquor than MHT users. Conclusions: Among postmenopausal women, the association between alcohol intake and parent oestrogen, but not oestrogen metabolite concentrations, may be influenced by MHT and type of alcohol.
Collapse
Affiliation(s)
- Mary C Playdon
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 6909 Medical Centre Drive, Rockville, MD 20850, USA
| | - Sally B Coburn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 6909 Medical Centre Drive, Rockville, MD 20850, USA
| | - Steven C Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 6909 Medical Centre Drive, Rockville, MD 20850, USA
| | - Louise A Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 6909 Medical Centre Drive, Rockville, MD 20850, USA
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 6909 Medical Centre Drive, Rockville, MD 20850, USA
| | - Garnet Anderson
- Public Health Sciences Division, University of Washington, 1100 Fairview Ave. N, M3-C102, Seattle, WA 98109, USA
| | - Robert Wallace
- College of Public Health, The University of Iowa, 145 N. Riverside drive, 100 CPHB, Iowa City, IA 52242, USA
| | - Roni T Falk
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 6909 Medical Centre Drive, Rockville, MD 20850, USA
| | - Ruth Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 6909 Medical Centre Drive, Rockville, MD 20850, USA
| | - Xia Xu
- Hormone Analysis Unit, Protein Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research Inc., P.O. Box B, Frederick, MD 21702-1201, USA
| | - Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 6909 Medical Centre Drive, Rockville, MD 20850, USA
| |
Collapse
|
32
|
Berstein LM, Berlev IV, Baltrukova AN. Endometrial cancer evolution: new molecular-biologic types and hormonal-metabolic shifts. Future Oncol 2017; 13:2593-2605. [PMID: 29168655 DOI: 10.2217/fon-2017-0217] [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] [Indexed: 12/17/2022] Open
Abstract
The question hidden in the title of this manuscript (whether the topic develops or remains constant) is important for all areas of science. It is also a serious problem for endometrial cancer (EC) study. In recent times the incidence of EC gradually increases in parallel with obesity epidemics. The main point of this review was evaluation of changes in EC area in last few decades, which are not only seen in tumor incidence, but also in its biology, hormonal-metabolic characteristics of patients and in the ratio of risk and anti-risk factors. One can hope that data accumulated recently and summarized here under the notion of EC evolution will find its use for advancement of EC prevention and treatment.
Collapse
Affiliation(s)
- Lev M Berstein
- Laboratory of Oncoendocrinology, NN Petrov Research Institute of Oncology, St Petersburg, Russia
| | - Igor V Berlev
- Department of Gynecological Oncology, NN Petrov Research Institute of Oncology, St Petersburg, Russia.,Department of Obstetrics and Gynecology, Northwestern State Medical University named after II Mechnikov, St Petersburg, Russia
| | - Alexandra N Baltrukova
- Laboratory of Oncoendocrinology, NN Petrov Research Institute of Oncology, St Petersburg, Russia.,Department of Obstetrics and Gynecology, Northwestern State Medical University named after II Mechnikov, St Petersburg, Russia
| |
Collapse
|
33
|
Meng X, Sun H, Yang L, Yin R, Qi L. A hydroxylated flavonol, fisetin inhibits the formation of a carcinogenic estrogen metabolite. Steroids 2017; 119:53-56. [PMID: 28119082 DOI: 10.1016/j.steroids.2017.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 12/22/2022]
Abstract
Fisetin can be found in a wide variety of plants and possesses strong efficacy against many cancers. 17β-Estradiol (E2) is hydrolyzed to 4-hydroxy-E2 (4-OHE2) via cytochrome P450 (CYP) 1B1 in vivo. In estrogen target tissues including the mammary gland, ovaries, and uterus, CYP1B1 is highly expressed, and 4-OHE2 is predominantly formed in cancerous tissues. Herein, we investigated the inhibitory activity of fisetin and flavone against CYP1B1 using estrogen E2 as substrate in vitro to reveal structure-activity relationship between structure of flavonoids and inhibition. The results showed that fisetin possessed inhibitory effect on CYP1B1 activity. Compared with flavone, the inhibition of fisetin was stronger. The Vmax and Ki values were 1.950±0.157pmol/μgprotein/min and 4.925±0.689nM for fisetin and 2.277±0.231pmol/μgprotein/min and 9.148±2.150nM for flavone, respectively. By kinetic analyses, both fisetin and flavone displayed mixed inhibition. Taken together the data suggested that fisetin is able to inhibit the formation of carcinogenic 4-OHE2 from E2, which reveals one of its anti-cancer mechanisms and helps to reveal the relationship between the structure of flavonoids and the inhibition CYP1B1 for discovering new drugs in cancer therapy and prevention.
Collapse
Affiliation(s)
- Xin Meng
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24 Heping Road, Harbin 150040, PR China.
| | - Hui Sun
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24 Heping Road, Harbin 150040, PR China
| | - Lianrong Yang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24 Heping Road, Harbin 150040, PR China
| | - Rui Yin
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24 Heping Road, Harbin 150040, PR China
| | - Lehui Qi
- School of Pharmacy, Heilongjiang University of Chinese Medicine, No. 24 Heping Road, Harbin 150040, PR China
| |
Collapse
|
34
|
Zang T, Tamae D, Mesaros C, Wang Q, Huang M, Blair IA, Penning TM. Simultaneous quantitation of nine hydroxy-androgens and their conjugates in human serum by stable isotope dilution liquid chromatography electrospray ionization tandem mass spectrometry. J Steroid Biochem Mol Biol 2017; 165:342-355. [PMID: 27531846 PMCID: PMC5146996 DOI: 10.1016/j.jsbmb.2016.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 08/04/2016] [Accepted: 08/08/2016] [Indexed: 12/19/2022]
Abstract
Castration resistant prostate cancer (CRPC), the fatal form of prostate cancer, remains androgen dependent despite castrate levels of circulating testosterone (T) and 5α-dihydrotestosterone (DHT). To investigate mechanisms by which the tumor can synthesize its own androgens and develop resistance to abiraterone acetate and enzalutamide, methods to measure a complete androgen profile are imperative. Here, we report the development and validation of a stable isotope dilution liquid chromatography electrospray ionization tandem mass spectrometric (SID-LC-ESI-MS/MS) method to quantify nine human hydroxy-androgens as picolinates, simultaneously with requisite specificity and sensitivity. In the established method, the fragmentation patterns of all nine hydroxy-androgen picolinates were identified, and [13C3]-5α-androstane-3α, 17β-diol and [13C3]-5α-androstane-3β, 17β-diol used as internal standards were synthesized enzymatically. Intra-day and inter-day precision and accuracy corresponds to the U.S. Food and Drug Administration Criteria for Bioanalytical Method Validation. The lower limit of quantitation (LLOQ) of nine hydroxy-androgens is 1.0pg to 2.5pg on column. Diols which have been infrequently measured: 5-androstene-3β, 17β-diol and 5α-androstane-3α, 17β-diol can be determined in serum at values as low as 1.0pg on column. The method also permits the quantitation of conjugated hydroxy-androgens following enzymatic digestion. While direct detection of steroid conjugates by electrospray-ionization tandem mass spectrometry has advantages the detection of unconjugated and conjugated steroids would require separate methods for each set of analytes. Our method was applied to pooled serum from male and female donors to provide reference values for both unconjugated and conjugated hydroxy-androgens. This method will allow us to interrogate the involvement of the conversion of 5-androstene-3β, 17β-diol to T, the backdoor pathway involving the conversion of 5α-androstane-3α, 17β-diol to DHT and the inactivation of DHT to 5α-androstane-3β, 17β-diol in advanced prostate cancer.
Collapse
Affiliation(s)
- Tianzhu Zang
- Center for Excellence in Environmental Toxicology, Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Daniel Tamae
- Center for Excellence in Environmental Toxicology, Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Clementina Mesaros
- Center for Excellence in Environmental Toxicology, Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Center for Cancer Pharmacology, Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Qingqing Wang
- Center for Cancer Pharmacology, Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Meng Huang
- Center for Excellence in Environmental Toxicology, Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Ian A Blair
- Center for Excellence in Environmental Toxicology, Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Center for Cancer Pharmacology, Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Trevor M Penning
- Center for Excellence in Environmental Toxicology, Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Center for Cancer Pharmacology, Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
35
|
Kitson SJ, Evans DG, Crosbie EJ. Identifying High-Risk Women for Endometrial Cancer Prevention Strategies: Proposal of an Endometrial Cancer Risk Prediction Model. Cancer Prev Res (Phila) 2016; 10:1-13. [DOI: 10.1158/1940-6207.capr-16-0224] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/20/2016] [Accepted: 11/21/2016] [Indexed: 11/16/2022]
|
36
|
Manna PR, Molehin D, Ahmed AU. Dysregulation of Aromatase in Breast, Endometrial, and Ovarian Cancers: An Overview of Therapeutic Strategies. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:487-537. [PMID: 27865465 DOI: 10.1016/bs.pmbts.2016.10.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aromatase is the rate-limiting enzyme in the biosynthesis of estrogens, which play crucial roles on a spectrum of developmental and physiological processes. The biological actions of estrogens are classically mediated by binding to two estrogen receptors (ERs), ERα and ERβ. Encoded by the cytochrome P450, family 19, subfamily A, polypeptide 1 (CYP19A1) gene, aromatase is expressed in a wide variety of tissues, as well as benign and malignant tumors, and is regulated in a pathway- and tissue-specific manner. Overexpression of aromatase, leading to elevated systemic levels of estrogen, is unequivocally linked to the pathogenesis and growth of a number malignancies, including breast, endometrium, and ovarian cancers. Aromatase inhibitors (AIs) are routinely used to treat estrogen-dependent breast cancers in postmenopausal women; however, their roles in endometrial and ovarian cancers remain obscure. While AI therapy is effective in hormone sensitive cancers, they diminish estrogen production throughout the body and, thus, generate undesirable side effects. Despite the effectiveness of AI therapy, resistance to endocrine therapy remains a major concern and is the leading cause of cancer death. Considerable advances, toward mitigating these issues, have evolved in conjunction with a number of histone deacetylase (HDAC) inhibitors for countering an assortment of diseases and cancers, including the aforesaid malignancies. HDACs are a family of enzymes that are frequently dysregulated in human tumors. This chapter will discuss the current understanding of aberrant regulation and expression of aromatase in breast, endometrial, and ovarian cancers, and potential therapeutic strategies for prevention and treatment of these life-threatening diseases.
Collapse
Affiliation(s)
- P R Manna
- Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, United States.
| | - D Molehin
- Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, United States
| | - A U Ahmed
- Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, United States
| |
Collapse
|
37
|
Brinton LA, Trabert B, Anderson GL, Falk RT, Felix AS, Fuhrman BJ, Gass ML, Kuller LH, Pfeiffer RM, Rohan TE, Strickler HD, Xu X, Wentzensen N. Serum Estrogens and Estrogen Metabolites and Endometrial Cancer Risk among Postmenopausal Women. Cancer Epidemiol Biomarkers Prev 2016; 25:1081-9. [PMID: 27197275 PMCID: PMC4930692 DOI: 10.1158/1055-9965.epi-16-0225] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 04/05/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Although endometrial cancer is clearly influenced by hormonal factors, few epidemiologic studies have investigated the role of endogenous estrogens or especially estrogen metabolites. METHODS We conducted a nested case-control study within the Women's Health Initiative Observational Study (WHI-OS), a cohort of 93,676 postmenopausal women recruited between 1993 and 1998. Using baseline serum samples from women who were non-current hormone users with intact uteri, we measured 15 estrogens/estrogen metabolites via HPLC/MS-MS among 313 incident endometrial cancer cases (271 type I, 42 type II) and 354 matched controls, deriving adjusted ORs and 95% confidence intervals (CI) for overall and subtype-specific endometrial cancer risk. RESULTS Parent estrogens (estrone and estradiol) were positively related to endometrial cancer risk, with the highest risk observed for unconjugated estradiol (OR 5th vs. 1st quintile = 6.19; 95% CI, 2.95-13.03, Ptrend = 0.0001). Nearly all metabolites were significantly associated with elevated risks, with some attenuation after adjustment for unconjugated estradiol (residual risks of 2- to 3-fold). Body mass index (kg/m(2), BMI) relations were somewhat reduced after adjustment for estrogen levels. The association with unconjugated estradiol was stronger for type I than type II tumors (Phet = 0.01). CONCLUSIONS Parent estrogens as well as individual metabolites appeared to exert generalized uterotropic activity, particularly for type I tumors. The effects of obesity on risk were only partially explained by estrogens. IMPACT These findings enhance our understanding of estrogen mechanisms involved in endometrial carcinogenesis but also highlight the need for studying additional markers that may underlie the effects on risk of certain risk factors, for example, obesity. Cancer Epidemiol Biomarkers Prev; 25(7); 1081-9. ©2016 AACR.
Collapse
Affiliation(s)
- Louise A Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland.
| | - Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Garnet L Anderson
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Roni T Falk
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Ashley S Felix
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland. Division of Epidemiology, The Ohio State University College of Public Health, Columbus, Ohio
| | - Barbara J Fuhrman
- Department of Epidemiology, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | - Lewis H Kuller
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Howard D Strickler
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Xia Xu
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
38
|
Trabert B, Brinton LA, Anderson GL, Pfeiffer RM, Falk RT, Strickler HD, Sliesoraitis S, Kuller LH, Gass ML, Fuhrman BJ, Xu X, Wentzensen N. Circulating Estrogens and Postmenopausal Ovarian Cancer Risk in the Women's Health Initiative Observational Study. Cancer Epidemiol Biomarkers Prev 2016; 25:648-56. [PMID: 26908437 PMCID: PMC4873440 DOI: 10.1158/1055-9965.epi-15-1272-t] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/09/2016] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Hormonal and reproductive factors contribute to the development of ovarian cancer, but few studies have examined associations between circulating estrogens and estrogen metabolites and ovarian cancer risk. We evaluated whether serum estrogens and estrogen metabolite levels are associated with ovarian cancer risk among postmenopausal women in a nested case-control study in the Women's Health Initiative (WHI) Observational Study (OS). METHODS We selected all 169 eligible epithelial ovarian cancer cases and 412 matched controls from women enrolled in WHI-OS who were not using menopausal hormones at baseline. Baseline levels of 15 estrogens and estrogen metabolites were measured via liquid chromatography/tandem mass spectrometry. Associations with ovarian cancer risk overall and stratified by histologic subtype (serous/nonserous) were analyzed using logistic regression. The mean time from serum collection to cancer diagnosis was 6.9 years. RESULTS Overall, we observed modest ovarian cancer risk associations among women with higher levels of estrone [OR (95% confidence interval) quintile (Q)5 vs. Q1: 1.54 (0.82-2.90), Ptrend = 0.05], as well as 2- and 4-methoxyestrone metabolites [2.03 (1.06-3.88), Ptrend = 0.02; 1.86 (0.98-3.56), Ptrend = 0.01, respectively]. Associations of estrogens and estrogen metabolites varied substantially by histologic subtype. Associations with serous tumors were universally null, while estrone [2.65 (1.09-6.45), Ptrend = 0.01, Pheterogeneity = 0.04], unconjugated estradiol [2.72 (1.04-7.14), Ptrend = 0.03, Pheterogeneity = 0.02] and many of the 2-, 4-, and 16-pathway metabolites were positively associated with nonserous tumors. CONCLUSIONS Our study provides novel molecular data showing an association of the parent estrogens and several estrogen metabolites with nonserous ovarian cancers. IMPACT These findings further support the heterogeneous etiology of ovarian cancer. Cancer Epidemiol Biomarkers Prev; 25(4); 648-56. ©2016 AACR.
Collapse
Affiliation(s)
- Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland.
| | - Louise A Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Garnet L Anderson
- Division of Public Health Sciences, Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Roni T Falk
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Howard D Strickler
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Sarunas Sliesoraitis
- Department of Hematology and Medical Oncology, University of Texas at San Antonio, San Antonio, Texas
| | - Lewis H Kuller
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Margery L Gass
- Women's Health Institute, Cleveland Clinic, Cleveland, Ohio
| | - Barbara J Fuhrman
- Department of Epidemiology, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Xia Xu
- Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|