1
|
Chumsri S. Serum hormone concentrations and individualisation of breast cancer prevention. Lancet Oncol 2024; 25:8-9. [PMID: 38181807 DOI: 10.1016/s1470-2045(23)00635-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2024]
Affiliation(s)
- Saranya Chumsri
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
2
|
Cuzick J, Chu K, Keevil B, Brentnall AR, Howell A, Zdenkowski N, Bonanni B, Loibl S, Holli K, Evans DG, Cummings S, Dowsett M. Effect of baseline oestradiol serum concentration on the efficacy of anastrozole for preventing breast cancer in postmenopausal women at high risk: a case-control study of the IBIS-II prevention trial. Lancet Oncol 2024; 25:108-116. [PMID: 38070530 DOI: 10.1016/s1470-2045(23)00578-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND An increased risk of breast cancer is associated with high serum concentrations of oestradiol and testosterone in postmenopausal women, but little is known about how these hormones affect response to endocrine therapy for breast cancer prevention or treatment. We aimed to assess the effects of serum oestradiol and testosterone concentrations on the efficacy of the aromatase inhibitor anastrozole for the prevention of breast cancer in postmenopausal women at high risk. METHODS In this case-control study we used data from the IBIS-II prevention trial, a randomised, controlled, double-blind trial in postmenopausal women aged 40-70 years at high risk of breast cancer, conducted in 153 breast cancer treatment centres across 18 countries. In the trial, women were randomly assigned (1:1) to receive anastrozole (1 mg/day, orally) or placebo daily for 5 years. In this pre-planned case-control study, the primary analysis was the effect of the baseline oestradiol to sex hormone binding globulin (SHBG) ratio (oestradiol-SHBG ratio) on the development of all breast cancers, including ductal carcinoma in situ (the primary endpoint in the trial). Cases were participants in whom breast cancer was reported after trial entry and until the cutoff on Oct 22, 2019, and who had valid blood samples and no use of hormone replacement therapy within 3 months of trial entry or during the trial. For each case, two controls without breast cancer were selected at random, matched on treatment group, age (within 2 years), and follow-up time (at least that of the matching case). For each treatment group, we applied a multinominal logistic regression likelihood-ratio trend test to assess what change in the proportion of cases was associated with a one-quartile change in hormone ratio. Controls were used only to determine quartile cutoffs. Profile likelihood 95% CIs were used to indicate the precision of estimates. A secondary analysis also investigated the effect of the baseline testosterone-SHBG ratio on breast cancer development. We also assessed relative benefit of anastrozole versus placebo (calculated as 1 - the ratio of breast cancer cases in the anastrozole group to cases in the placebo group). The trial was registered with ISRCTN (number ISRCTN31488319) and completed recruitment on Jan 31, 2012, but long-term follow-up is ongoing. FINDINGS 3864 women were recruited into the trial between Feb 2, 2003, and Jan 31, 2012, and randomly assigned to receive anastrozole (n=1920) or placebo (n=1944). Median follow-up time was 131 months (IQR 106-156), during which 85 (4·4%) cases of breast cancer in the anastrozole group and 165 (8·5%) in the placebo group were identified. No data on gender, race, or ethnicity were collected. After exclusions, the case-control study included 212 participants from the anastrozole group (72 cases, 140 controls) and 416 from the placebo group (142 cases, 274 controls). A trend of increasing breast cancer risk with increasing oestradiol-SHBG ratio was found in the placebo group (trend per quartile 1·25 [95% CI 1·08 to 1·45], p=0·0033), but not in the anastrozole group (1·06 [0·86 to 1·30], p=0·60). A weaker effect was seen for the testosterone-SHBG ratio in the placebo group (trend 1·21 [1·05 to 1·41], p=0·011), but again not in the anastrozole group (trend 1·18 [0·96 to 1·46], p=0·11). A relative benefit of anastrozole was seen in quartile 2 (0·55 [95% CI 0·13 to 0·78]), quartile 3 (0·54 [0·22 to 0·74], and quartile 4 (0·56 [0·23 to 0·76]) of oestradiol-SHBG ratio, but not in quartile 1 (0·18 [-0·60 to 0·59]). INTERPRETATION These results suggest that serum hormones should be measured more routinely and integrated into risk management decisions. Measuring serum hormone concentrations is inexpensive and might help clinicians differentiate which women will benefit most from an aromatase inhibitor. FUNDING Cancer Research UK, National Health and Medical Research Council (Australia), Breast Cancer Research Foundation, and DaCosta Fund.
Collapse
Affiliation(s)
- Jack Cuzick
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK.
| | - Kim Chu
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Brian Keevil
- University South Manchester NHS Foundation Trust, Manchester, UK
| | - Adam R Brentnall
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Anthony Howell
- Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| | - Nicholas Zdenkowski
- Faculty of Health and Medicine, University of Newcastle, Newcastle, Australia
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, European Institute of Oncology IRCCS, Milan, Italy
| | - Sibylle Loibl
- German Breast Group, Goethe University of Frankfurt, Frankfurt, Germany
| | | | - D Gareth Evans
- Centre for Genomic Medicine, University of Manchester, Manchester, UK
| | - Steve Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Mitch Dowsett
- Institute of Cancer Research, Royal Marsden Hospital, London, UK
| |
Collapse
|
3
|
Shalabi SF, LaBarge MA. Cellular and molecular mechanisms of breast cancer susceptibility. Clin Sci (Lond) 2022; 136:1025-1043. [PMID: 35786748 DOI: 10.1042/cs20211158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 06/16/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022]
Abstract
There is a plethora of recognized risk factors for breast cancer (BC) with poorly understood or speculative biological mechanisms. The lack of prevention options highlights the importance of understanding the mechanistic basis of cancer susceptibility and finding new targets for breast cancer prevention. Until now, we have understood risk and cancer susceptibility primarily through the application of epidemiology and assessing outcomes in large human cohorts. Relative risks are assigned to various human behaviors and conditions, but in general the associations are weak and there is little understanding of mechanism. Aging is by far the greatest risk factor for BC, and there are specific forms of inherited genetic risk that are well-understood to cause BC. We propose that bringing focus to the biology underlying these forms of risk will illuminate biological mechanisms of BC susceptibility.
Collapse
Affiliation(s)
- Sundus F Shalabi
- Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, CA, U.S.A
- Medical Research Center, Al-Quds University, Jerusalem, Palestine
| | - Mark A LaBarge
- Department of Population Sciences, Beckman Research Institute, City of Hope, Duarte, CA, U.S.A
- Center for Cancer and Aging, Beckman Research Institute, City of Hope, Duarte, CA, U.S.A
- Center for Cancer Biomarkers Research (CCBIO), Bergen, Norway
| |
Collapse
|
4
|
Mohan A, Kumar V, Brahmachari S, Pandya B. A Study on Clinico-Pathological Profile of Breast Cancer Patients and Their Correlation With Uterine Fibroids Using Hormone Level and Receptor Status Assessment. BREAST CANCER: BASIC AND CLINICAL RESEARCH 2022; 16:11782234221090197. [PMID: 35462755 PMCID: PMC9019335 DOI: 10.1177/11782234221090197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/08/2022] [Indexed: 11/27/2022] Open
Abstract
Purpose: To study the clinico-pathological profile of breast cancer patients and the prevalence of uterine fibroids in them, their hormonal levels and hormone receptor status. Patients and methods: 52 patients with breast cancer who attended AIIMS Bhopal from November 2018 to January 2020 were selected, with their clinical details, triple assessment and other investigations for further management being performed and recorded. The presence of uterine fibroids was assessed using ultrasound of the abdomen, and for patients who had undergone hysterectomy, previous medical records were examined to ascertain the history of uterine fibroids. Serum levels of estrogen and progesterone were assessed using chemi-luminescent micro-particle immune assay (CMIA). Results: The mean age of patients was 50.35 ± 10.87 years. 36.54% of our patients had uterine fibroids, of whom 15.38% had undergone hysterectomy for the same, and 21.15% was detected on ultrasound of the abdomen during evaluation. Among patients with uterine fibroids, 84.2% were hormone receptor-positive, while in patients without uterine fibroids, only 57.6% had positive receptors. (P = 0.049). Among premenopausal patients, there was a statistically significant difference in serum progesterone values between patients with and without uterine fibroids. Conclusion: The prevalence of uterine fibroids in our study group of breast cancer patients was found to be high. The role of estrogen and progesterone in the pathophysiology of both diseases and the common risk factors involved may biologically explain this finding. Breast cancer and other estrogen associated disorders may hold future research prospects.
Collapse
Affiliation(s)
- Anjaly Mohan
- Department of Surgical Oncology, All India Institute of Medical Sciences, Bhopal, India
| | - Vinay Kumar
- Department of Surgical Oncology, All India Institute of Medical Sciences, Bhopal, India
| | - Swagata Brahmachari
- Department of General Surgery, All India Institute of Medical Sciences, Bhopal, India
| | - Bharati Pandya
- Department of General Surgery, All India Institute of Medical Sciences, Bhopal, India
| |
Collapse
|
5
|
Drummond AE, Swain CTV, Brown KA, Dixon-Suen SC, Boing L, van Roekel EH, Moore MM, Gaunt TR, Milne RL, English DR, Martin RM, Lewis SJ, Lynch BM. Linking Physical Activity to Breast Cancer via Sex Steroid Hormones, Part 2: The Effect of Sex Steroid Hormones on Breast Cancer Risk. Cancer Epidemiol Biomarkers Prev 2022; 31:28-37. [PMID: 34670801 PMCID: PMC7612577 DOI: 10.1158/1055-9965.epi-21-0438] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/10/2021] [Accepted: 10/07/2021] [Indexed: 11/25/2022] Open
Abstract
We undertook a systematic review and appraised the evidence for an effect of circulating sex steroid hormones and sex hormone-binding globulin (SHBG) on breast cancer risk in pre- and postmenopausal women. Systematic searches identified prospective studies relevant to this review. Meta-analyses estimated breast cancer risk for women with the highest compared with the lowest level of sex hormones, and the DRMETA Stata package was used to graphically represent the shape of these associations. The ROBINS-E tool assessed risk of bias, and the GRADE system appraised the strength of evidence. In premenopausal women, there was little evidence that estrogens, progesterone, or SHBG were associated with breast cancer risk, whereas androgens showed a positive association. In postmenopausal women, higher estrogens and androgens were associated with an increase in breast cancer risk, whereas higher SHBG was inversely associated with risk. The strength of the evidence quality ranged from low to high for each hormone. Dose-response relationships between sex steroid hormone concentrations and breast cancer risk were most notable for postmenopausal women. These data support the plausibility of a role for sex steroid hormones in mediating the causal relationship between physical activity and the risk of breast cancer.See related reviews by Lynch et al., p. 11 and Swain et al., p. 16.
Collapse
Affiliation(s)
- Ann E Drummond
- Cancer Epidemiology Division, Cancer Council Victoria, Victoria, Australia
| | | | - Kristy A Brown
- Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Suzanne C Dixon-Suen
- Cancer Epidemiology Division, Cancer Council Victoria, Victoria, Australia
- Institute for Physical Activity and Nutrition, Deakin University, Geelong, Victoria, Australia
| | - Leonessa Boing
- Laboratory of Research in Leisure and Physical Activity, Santa Catarina State University, Florianópolis, Brazil
| | - Eline H van Roekel
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Melissa M Moore
- Medical Oncology, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Tom R Gaunt
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia
| | - Dallas R English
- Cancer Epidemiology Division, Cancer Council Victoria, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Richard M Martin
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
- NIHR Biomedical Research Centre at University Hospitals Bristol and Weston NHS Foundation Trust and the University of Bristol, Bristol, United Kingdom
| | - Sarah J Lewis
- Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Brigid M Lynch
- Cancer Epidemiology Division, Cancer Council Victoria, Victoria, Australia.
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
6
|
Hegde M, Joshi MB. Comprehensive analysis of regulation of DNA methyltransferase isoforms in human breast tumors. J Cancer Res Clin Oncol 2021; 147:937-971. [PMID: 33604794 PMCID: PMC7954751 DOI: 10.1007/s00432-021-03519-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/10/2021] [Indexed: 12/14/2022]
Abstract
Significant reprogramming of epigenome is widely described during pathogenesis of breast cancer. Transformation of normal cell to hyperplastic cell and to neoplastic phenotype is associated with aberrant DNA (de)methylation, which, through promoter and enhancer methylation changes, activates oncogenes and silence tumor suppressor genes in variety of tumors including breast. DNA methylation, one of the major epigenetic mechanisms is catalyzed by evolutionarily conserved isoforms namely, DNMT1, DNMT3A and DNMT3B in humans. Over the years, studies have demonstrated intricate and complex regulation of DNMT isoforms at transcriptional, translational and post-translational levels. The recent findings of allosteric regulation of DNMT isoforms and regulation by other interacting chromatin modifying proteins emphasizes functional integrity and their contribution for the development of breast cancer and progression. DNMT isoforms are regulated by several intrinsic and extrinsic parameters. In the present review, we have extensively performed bioinformatics analysis of expression of DNMT isoforms along with their transcriptional and post-transcriptional regulators such as transcription factors, interacting proteins, hormones, cytokines and dietary elements along with their significance during pathogenesis of breast tumors. Our review manuscript provides a comprehensive understanding of key factors regulating DNMT isoforms in breast tumor pathology and documents unsolved issues.
Collapse
Affiliation(s)
- Mangala Hegde
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Planetarium Complex, Manipal, 576104, India
| | - Manjunath B Joshi
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Planetarium Complex, Manipal, 576104, India.
| |
Collapse
|
7
|
Park SJ, Kim TS, Park KH, Kwon WS, Kim JJ. Serum concentration of sex hormone-binding globulin in healthy volunteers and patients with breast cancer stratified by sex and age. Oncol Lett 2020; 20:364-372. [PMID: 32565962 PMCID: PMC7285803 DOI: 10.3892/ol.2020.11549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/05/2020] [Indexed: 11/07/2022] Open
Abstract
The objective of the present study was to compare sex hormone-binding globulin (SHBG) levels according to sex (healthy male and female volunteers) and age to determine reference values. Serum SHBG expression levels in patients with breast cancer with different tumor burden states were also determined. A total of 109 samples were obtained from 34 patients in 3 different disease states (non-tumor, localized tumor and systemic metastasis) during follow-up. A sandwich ELISA was conducted to measure SHBG, cancer antigen (CA)15-3 and CA125 expression levels. Wilcoxon rank-sum tests were performed on non-normally distributed data and an unpaired t-test was used for normally distributed variables. SHBG expression levels were higher in females compared with males (P<0.0001). When SHBG expression levels were compared by sex, the difference was maintained in the age groups <30, 30–39 and ≥50 years, but not in the 40–49 years group. In males, SHBG expression levels increased until the age of 49 and then decreased (P=0.01). In females, SHBG expression levels exhibited a decreased trend until the age of 49 (P=0.66). In patients with breast cancer, the SHBG expression levels revealed a decreasing trend after the age of 50, which was different compared with the healthy females. There was a decreasing trend of SHBG expression levels from pre-menopause to post-menopause healthy volunteers (P=0.74). CA15-3 (r2=0.07; P=0.59) and CA 125 (r2=−0.18; P=0.17) levels did not exhibit any significant correlation with SHBG expression levels. There was a significant difference in the SHBG expression levels between male and female healthy volunteers. SHBG expression levels also revealed different patterns between healthy female volunteers and female patients with breast cancer ≥50 years of age. The present study demonstrated that SHBG does not have value as a biomarker, but different reference values according to age and sex may aid in predicting high-risk groups for hormone-dependent cancer and guide treatment direction for post-menopausal breast cancer.
Collapse
Affiliation(s)
- Se Jung Park
- Song Dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Tae Soo Kim
- Song Dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kyu Hyun Park
- Song Dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Woo Sun Kwon
- Song Dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jin Ju Kim
- Department of Laboratory Medicine, Inha University College of Medicine, Incheon 22332, Republic of Korea
| |
Collapse
|
8
|
Carvalho MJ, Subtil S, Rodrigues Â, Oliveira J, Figueiredo-Dias M. Controversial association between polycystic ovary syndrome and breast cancer. Eur J Obstet Gynecol Reprod Biol 2019; 243:125-132. [PMID: 31693949 DOI: 10.1016/j.ejogrb.2019.10.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 09/07/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022]
Abstract
Polycystic ovary syndrome (PCOS) risk factors overlap with breast cancer, and the hormonal profile may be implicated in breast cancer pathogenesis. This study aims to report a literature review considering epidemiological and molecular mechanisms that correlate PCOS and breast cancer, as well as the influence of PCOS treatment on the incidence of breast cancer. Epidemiological studies failed to adjust potential variables that affect the risk and have thus provided inconclusive results. Molecular effects of androgenic pathways in breast cancer have been studied and androgens seem to have an inhibitory effect on mammary epithelial proliferation. However, increased bioavailable androgens were associated with recurrence of breast cancer due to conversion to oestrogens. Sex hormone-binding globulin has a role in hormone-dependent cancers and can be considered a marker for PCOS; a gene profile has already been linked to breast cancer risk in these patients. PCOS medical treatment is a promising tool for stratifying breast cancer risk due to the metabolic influence and hormonal environment. Clinical reports are inconsistent, emphasizing the need for further studies with a prospective design. In the future, the role of pharmacological interventions in PCOS will increase knowledge and awareness of breast cancer pathogenesis and will help to refine breast cancer risk stratification.
Collapse
Affiliation(s)
- Maria João Carvalho
- University Clinic of Gynaecology, Faculty of Medicine, University of Coimbra, Clinical Academic Centre of Coimbra, CACC, Coimbra, Portugal; Gynaecology Service, Coimbra Hospital and University Centre, Coimbra, Portugal.
| | - Simone Subtil
- University Clinic of Gynaecology, Faculty of Medicine, University of Coimbra, Clinical Academic Centre of Coimbra, CACC, Coimbra, Portugal; Gynaecology Service, Coimbra Hospital and University Centre, Coimbra, Portugal
| | - Ângela Rodrigues
- University Clinic of Gynaecology, Faculty of Medicine, University of Coimbra, Clinical Academic Centre of Coimbra, CACC, Coimbra, Portugal; Gynaecology Service, Coimbra Hospital and University Centre, Coimbra, Portugal
| | - Joana Oliveira
- University Clinic of Gynaecology, Faculty of Medicine, University of Coimbra, Clinical Academic Centre of Coimbra, CACC, Coimbra, Portugal; Gynaecology Service, Coimbra Hospital and University Centre, Coimbra, Portugal
| | - Margarida Figueiredo-Dias
- University Clinic of Gynaecology, Faculty of Medicine, University of Coimbra, Clinical Academic Centre of Coimbra, CACC, Coimbra, Portugal; Gynaecology Service, Coimbra Hospital and University Centre, Coimbra, Portugal
| |
Collapse
|
9
|
Thompson PA, Preece C, Stopeck AT. Breast Cancer Prevention. FUNDAMENTALS OF CANCER PREVENTION 2019:543-606. [DOI: 10.1007/978-3-030-15935-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
10
|
La Vignera S, Condorelli RA, Cannarella R, Duca Y, Calogero AE. Sport, doping and female fertility. Reprod Biol Endocrinol 2018; 16:108. [PMID: 30449281 PMCID: PMC6241032 DOI: 10.1186/s12958-018-0437-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 10/24/2018] [Indexed: 01/02/2023] Open
Abstract
This article is a review that addresses the following topics, divided by paragraphs. The first paragraph investigates the effects of physical activity on ovarian function, analyzing in particular the changes concerning the serum concentrations of follicle-stimulating hormone, luteinizing hormone, prolactin, growth hormone, thyroid hormones, leptin, ghrelin, neuropeptide Y. The second paragraph analyzes the effects of doping on the hypothalamic-pituitary-ovarian axis. Finally, the last paragraph analyzes the PCOS category, evaluating the effects of hyperandrogenism in relation to athletic performance.
Collapse
Affiliation(s)
- Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", via S. Sofia 78, 95123, Catania, Italy.
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", via S. Sofia 78, 95123, Catania, Italy
| | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", via S. Sofia 78, 95123, Catania, Italy
| | - Ylenia Duca
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", via S. Sofia 78, 95123, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", via S. Sofia 78, 95123, Catania, Italy
| |
Collapse
|
11
|
Cao A, Huang L, Shao Z. The Preventive Intervention of Hereditary Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1026:41-57. [PMID: 29282679 DOI: 10.1007/978-981-10-6020-5_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Approximately 5-10% of breast cancer is considered to be hereditary. Familial breast cancers exhibit a dominant hereditary pattern, which typically have an early age of onset and are accompanied by symptoms of ovarian cancer, bilateral breast cancer, or male breast cancer. BRCA gene mutation carriers should be regarded as high-risk groups for breast cancer, which necessitates early examination of breast cancer. Studies have built up kinds of predictive models and recommended that female BRCA mutation carriers should receive breast self-test training and take monthly breast self-examination. Familial or hereditary breast cancer family members are high-risk groups, and their risks of breast cancer can be reduced by chemoprevention, including dietary composition adjustment and application of endocrine drugs. In recent years, large-scale clinical trials have shown the important role of chemoprevention in reducing the occurrence of hereditary breast cancer. Prophylactic mastectomy is also suitable for healthy women with high breast cancer risk factors. It can reduce the incidence rate of breast cancer in high-risk women by 90% and decrease the breast cancer mortality rate in medium-risk and high-risk women by 100% and 81%, respectively.
Collapse
Affiliation(s)
- Ayong Cao
- Department of Breast Surgery, Shanghai Cancer Center/Cancer Institute, Fudan University, No.270 Dong'an Road, Shanghai, China
| | - Liang Huang
- Department of Breast Surgery, Shanghai Cancer Center/Cancer Institute, Fudan University, No.270 Dong'an Road, Shanghai, China
| | - Zhimin Shao
- Department of Breast Surgery, Shanghai Cancer Center/Cancer Institute, Fudan University, No.270 Dong'an Road, Shanghai, China.
| |
Collapse
|
12
|
Shieh Y, Hu D, Ma L, Huntsman S, Gard CC, Leung JWT, Tice JA, Ziv E, Kerlikowske K, Cummings SR. Joint relative risks for estrogen receptor-positive breast cancer from a clinical model, polygenic risk score, and sex hormones. Breast Cancer Res Treat 2017; 166:603-612. [PMID: 28791495 DOI: 10.1007/s10549-017-4430-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/29/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Models that predict the risk of estrogen receptor (ER)-positive breast cancers may improve our ability to target chemoprevention. We investigated the contributions of sex hormones to the discrimination of the Breast Cancer Surveillance Consortium (BCSC) risk model and a polygenic risk score comprised of 83 single nucleotide polymorphisms. METHODS We conducted a nested case-control study of 110 women with ER-positive breast cancers and 214 matched controls within a mammography screening cohort. Participants were postmenopausal and not on hormonal therapy. The associations of estradiol, estrone, testosterone, and sex hormone binding globulin with ER-positive breast cancer were evaluated using conditional logistic regression. We assessed the individual and combined discrimination of estradiol, the BCSC risk score, and polygenic risk score using the area under the receiver operating characteristic curve (AUROC). RESULTS Of the sex hormones assessed, estradiol (OR 3.64, 95% CI 1.64-8.06 for top vs bottom quartile), and to a lesser degree estrone, was most strongly associated with ER-positive breast cancer in unadjusted analysis. The BCSC risk score (OR 1.32, 95% CI 1.00-1.75 per 1% increase) and polygenic risk score (OR 1.58, 95% CI 1.06-2.36 per standard deviation) were also associated with ER-positive cancers. A model containing the BCSC risk score, polygenic risk score, and estradiol levels showed good discrimination for ER-positive cancers (AUROC 0.72, 95% CI 0.65-0.79), representing a significant improvement over the BCSC risk score (AUROC 0.58, 95% CI 0.50-0.65). CONCLUSION Adding estradiol and a polygenic risk score to a clinical risk model improves discrimination for postmenopausal ER-positive breast cancers.
Collapse
Affiliation(s)
- Yiwey Shieh
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, Box 0320, 1545 Divisadero Street, San Francisco, CA, 94115, USA.
| | - Donglei Hu
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, Box 0320, 1545 Divisadero Street, San Francisco, CA, 94115, USA
| | - Lin Ma
- University of California, San Francisco, Box 1793, 550 16th Street, San Francisco, CA, 94158, USA
| | - Scott Huntsman
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, Box 0320, 1545 Divisadero Street, San Francisco, CA, 94115, USA
| | - Charlotte C Gard
- Department of Economics, Applied Statistics, and International Business, New Mexico State University, MSC 3CQ, P.O. Box 30001, Las Cruces, NM, 88003, USA
| | - Jessica W T Leung
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1350, Houston, TX, 77030, USA
| | - Jeffrey A Tice
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, Box 0320, 1545 Divisadero Street, San Francisco, CA, 94115, USA
| | - Elad Ziv
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, Box 0320, 1545 Divisadero Street, San Francisco, CA, 94115, USA
| | - Karla Kerlikowske
- Department of Medicine and Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA.,General Internal Medicine Section, San Francisco Veterans Affairs Medical Center, 4150 Clement St, Mailing Code 111A1, San Francisco, CA, 94121, USA
| | - Steven R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, Box 0560, 550 16th Street, 2nd Floor, San Francisco, CA, 94159, USA
| |
Collapse
|
13
|
Di Lascio S, Pagani O. New insights into endocrine therapy for young women with breast cancer. ACTA ACUST UNITED AC 2015; 11:343-54. [PMID: 26102472 DOI: 10.2217/whe.15.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Managing estrogen receptor-positive breast cancer in young women (<40 years) requires a multidisciplinary/personalized approach, covering both clinical and psychosocial aspects. Five years of tamoxifen has been the standard adjuvant endocrine therapy for many years. Recent data from the adjuvant randomized trials TEXT-SOFT show that the aromatase inhibitor exemestane plus ovarian suppression significantly reduces recurrences as compared with tamoxifen plus ovarian suppression. The ATLAS and aTToM trials represent the first evidence of a beneficial effect of extended endocrine therapy with tamoxifen in premenopausal women. Outside of a clinical trial, no data support neoadjuvant endocrine therapy in young women. In the metastatic setting, tamoxifen or aromatase inhibitors, both with ovarian suppression/ablation, should be the preferred choice, unless rapid tumor shrinkage is needed. No data are available with fulvestrant in young patients.
Collapse
Affiliation(s)
- Simona Di Lascio
- Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland.,Breast Unit of Southern Switzerland (CSSI), Bellinzona, Switzerland
| | - Olivia Pagani
- Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland.,Breast Unit of Southern Switzerland (CSSI), Bellinzona, Switzerland
| |
Collapse
|
14
|
Tzeng HE, Muo CH, Chen HT, Hwang WL, Hsu HC, Tsai CH. Tamoxifen use reduces the risk of osteoporotic fractures in women with breast cancer in Asia: a nationwide population-based cohort study. BMC Musculoskelet Disord 2015; 16:123. [PMID: 25989902 PMCID: PMC4438570 DOI: 10.1186/s12891-015-0580-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/12/2015] [Indexed: 11/19/2022] Open
Abstract
Purpose Bone mineral density changes with tamoxifen treatment have been reported in pre- and post-menopausal women with breast cancer. However, there remains controversy as to whether tamoxifen significantly reduces fracture rates in different age groups. Breast cancer occurs at 10-20 years younger in Asian women compared with Western women. Therefore we conducted this population-based case-control study to determine whether or not tamoxifen use is associated with osteoporotic fractures. Patients and methods We selected 75488 women with breast cancer with no prior history of fractures from the Longitudinal Health Insurance Database for Catastrophic Illness Patients in 2000-2011. They were followed from the date of the diagnosis of breast cancer to the date a hip, vertebral or wrist fracture occurred. Because the use of tamoxifen was a time-dependent variable, we used a Cox proportional hazard model with time-dependent exposure covariates to estimate the risk of a fracture. Results There were 50257 and 25231 women with breast cancer who did and did not receive tamoxifen treatment, respectively. The tamoxifen users had lower risks for overall fractures with hazard ratios (HRs) of 0.52 and 0.59 in the crude and adjusted models (95 % CI = 0.45-0.61 and 0.51-0.69), respectively. They also had lower risks for hip (HR = 0.55, 95 % CI = 0.45-0.67) and vertebral (HR = 0.64, 95 % CI = 0.50-0.82) fractures in the adjusted model. The risk of fractures decreased with an increasing dosage of tamoxifen. Regardless of the age group, the tamoxifen users had a lower risk of fractures than the non-users. Conclusion In this Asian population-based case-control study, tamoxifen use was associated with a reduction in osteoporotic fractures, especially in hip fractures.
Collapse
Affiliation(s)
- Huey-En Tzeng
- Graduate Institute of Clinical Medicine, China Medical University, #91 Hsueh-Shih Road, Taichung, 404, Taiwan. .,Division of Hematology/Oncology, Taichung Veterans General Hospital, Taichung, Taiwan. .,School of Medicine, China Medical University, Taichung, Taiwan.
| | - Chih-Hsin Muo
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan.
| | - Hsien-Te Chen
- Department of Orthopedics, China Medical University Hospital, Taichung, Taiwan. .,School of Chinese Medicine, China Medical University, Taichung, Taiwan.
| | - Wen-Li Hwang
- Division of Hematology/Oncology, Taichung Veterans General Hospital, Taichung, Taiwan.
| | - Horng-Chang Hsu
- School of Medicine, China Medical University, Taichung, Taiwan. .,Department of Orthopedics, China Medical University Hospital, Taichung, Taiwan.
| | - Chun-Hao Tsai
- Graduate Institute of Clinical Medicine, China Medical University, #91 Hsueh-Shih Road, Taichung, 404, Taiwan. .,School of Medicine, China Medical University, Taichung, Taiwan. .,Department of Orthopedics, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
15
|
Davis SR, Worsley R. Androgen treatment of postmenopausal women. J Steroid Biochem Mol Biol 2014; 142:107-14. [PMID: 23727129 DOI: 10.1016/j.jsbmb.2013.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 04/12/2013] [Accepted: 05/18/2013] [Indexed: 01/23/2023]
Abstract
Testosterone is physiologically important for women. Serum testosterone levels decline with age, with the most precipitous fall being prior to menopause. There is no level of testosterone which defines a woman as being testosterone deficient. However, there is substantial high quality evidence to support the use of testosterone for the treatment of hypoactive sexual desire disorder in postmenopausal women. Although preliminary data suggests testosterone has favorable effects on bone and muscle mass, cognitive function and the cardiovascular system, further research regarding its therapeutic effects in these domains is warranted. As no testosterone product has been approved for women there is extensive off-label prescribing of testosterone products for women as well as the prescription of compounded therapy. This raises serious safety concerns and together with the evidence for the negative impact of FSD on quality of life, highlights the need for an approved testosterone formulation for women. This article is part of a Special Issue entitled 'Menopause'.
Collapse
Affiliation(s)
- Susan R Davis
- Women's Health Research Program, School of Public Health and Preventive Medicine, Department of Epidemiology and Preventive Medicine, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia.
| | - Roisin Worsley
- Women's Health Research Program, School of Public Health and Preventive Medicine, Department of Epidemiology and Preventive Medicine, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| |
Collapse
|
16
|
Samavat H, Kurzer MS. Estrogen metabolism and breast cancer. Cancer Lett 2014; 356:231-43. [PMID: 24784887 DOI: 10.1016/j.canlet.2014.04.018] [Citation(s) in RCA: 239] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 04/01/2014] [Accepted: 04/19/2014] [Indexed: 01/18/2023]
Abstract
There is currently accumulating evidence that endogenous estrogens play a critical role in the development of breast cancer. Estrogens and their metabolites have been studied in both pre- and postmenopausal women with more consistent results shown in the latter population, in part because of large hormonal variations during the menstrual cycle and far fewer studies having been performed in premenopausal women. In this review we describe in detail estrogen metabolism and associated genetic variations, and provide a critical review of the current literature regarding the role of estrogens and their metabolites in breast cancer risk.
Collapse
Affiliation(s)
- Hamed Samavat
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN, USA
| | - Mindy S Kurzer
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN, USA.
| |
Collapse
|
17
|
Farhat GN, Parimi N, Chlebowski RT, Manson JE, Anderson G, Huang AJ, Vittinghoff E, Lee JS, Lacroix AZ, Cauley JA, Jackson R, Grady D, Lane DS, Phillips L, Simon MS, Cummings SR. Sex hormone levels and risk of breast cancer with estrogen plus progestin. J Natl Cancer Inst 2013; 105:1496-503. [PMID: 24041978 DOI: 10.1093/jnci/djt243] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Although high endogenous sex hormone levels and estrogen plus progestin (E+P) therapy are associated with increased breast cancer risk, it is unknown whether pretreatment levels of sex hormones modify E+P effect on breast cancer. METHODS We conducted a nested case-control study within the Women's Health Initiative randomized clinical trial of E+P. The trial enrolled 16608 postmenopausal women aged 50 to 79 years with intact uterus and no breast cancer history. During a mean of 5.6 years of follow-up, 348 incident breast cancer case subjects were identified and matched with 348 control subjects. Case and control subjects had their sex hormone levels measured at baseline (estrogens, testosterone, progesterone, and sex hormone-binding globulin [SHBG]) and year 1 (estrogens and SHBG) using sensitive assays. All statistical tests were two-sided. RESULTS Statistically significant elevations in breast cancer risk were seen with greater pretreatment levels of total estradiol (P trend = .04), bioavailable estradiol (P trend = .03), estrone (P trend = .007), and estrone sulfate (P trend = .007). E+P increased all measured estrogens and SHGB at year 1 (all P < .001). The effect of E+P on breast cancer risk was strongest in women whose pretreatment levels of total estradiol, bioavailable estradiol, and estrone were in the lowest quartiles. For example, the odds ratio for E+P relative to placebo was 2.47 (95% confidence interval [CI] = 1.28 to 4.79) in the lowest total estradiol quartile, compared with 0.96 (95% CI = 0.44 to 2.09) in the highest total estradiol quartile; P interaction = .04). CONCLUSIONS Women with lower pr-treatment endogenous estrogen levels were at greater risk of breast cancer during E+P therapy compared with those with higher levels. Further studies are warranted to confirm these findings.
Collapse
Affiliation(s)
- Ghada N Farhat
- Affiliations of authors: San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA (GNF, NP, SRC); Faculty of Health Sciences, University of Balamand, Beirut, Lebanon (GNF); Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA (RTC); Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (JEM); Fred Hutchinson Cancer Research Center, Seattle, WA (GA, AZL); Department of Internal Medicine, University of California-San Francisco, San Francisco, CA (AH, DG); Department of Epidemiology and Biostatistics, University of California-San Francisco, San Francisco, CA (EV); Department of Internal Medicine, University of California-Davis, Sacramento, CA (JSL); Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA (JAC); Department of Internal Medicine, Ohio State University, Columbus, OH (RJ); Department of Preventive Medicine, Stony Brook University, Stony Brook, NY (DL); Department of Medicine, Emory University, Atlanta, GA (LP); Karmanos Cancer Institute, Wayne State University, Detroit, MI (MSS)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Christinat A, Di Lascio S, Pagani O. Hormonal therapies in young breast cancer patients: when, what and for how long? J Thorac Dis 2013; 5 Suppl 1:S36-46. [PMID: 23819026 DOI: 10.3978/j.issn.2072-1439.2013.05.25] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 05/19/2013] [Indexed: 12/26/2022]
Abstract
Breast cancer in young women (<40 years) is a rare and complex clinical and psychosocial condition, which deserves multidisciplinary and personalized approaches. In young women with hormone-receptor positive disease, 5 years of adjuvant tamoxifen, with or without ovarian suppression/ablation, is considered the standard endocrine therapy. The definitive role of adjuvant aromatase inhibitors has still to be elucidated: the upcoming results of the Tamoxifen and EXemestane Trial (TEXT) and Suppression of Ovarian Function Trial (SOFT) trials will help understanding if we can widen our current endocrine therapeutic options. The optimal duration of adjuvant endocrine therapy in young women also remains an unresolved issue. The recently reported results of the ATLAS and aTToM trials represent the first evidence of a beneficial effect of extended endocrine therapy in premenopausal women and provide an important opportunity in high-risk young patients. In the metastatic setting, endocrine therapy should be the preferred choice for endocrine responsive disease, unless there is evidence of endocrine resistance or need for rapid disease and/or symptom control. Tamoxifen in combination with ovarian suppression/ablation remains the 1st-line endocrine therapy of choice. Aromatase inhibitors in combination with ovarian suppression/ablation can be considered after progression on tamoxifen and ovarian suppression/ablation. Fulvestrant has not yet been studied in pre-menopausal women. Specific age-related treatment side effects (i.e., menopausal symptoms, change in body image and weight gain, cognitive function impairment, fertility damage/preservation, long-term organ dysfunction, sexuality) and the social impact of diagnosis and treatment (i.e., job discrimination, family management) should be carefully addressed when planning long-lasting endocrine therapies in young women with hormone-receptor positive early and advanced breast cancer.
Collapse
Affiliation(s)
- Alexandre Christinat
- Institute of Oncology of Southern Switzerland (IOSI) and Breast Unit of Southern Switzerland (CSSI), Bellinzona, Switzerland
| | | | | |
Collapse
|
19
|
Abstract
Testosterone is increasingly used as part of postmenopausal HRT regimens. Unfortunately, few androgenic preparations designed specifically for use in women have been approved by regulatory authorities. Ongoing concerns exist surrounding the potential long-term effects of testosterone therapy. Here, we review the most recent data on postmenopausal testosterone therapy, focusing particularly on the effects of testosterone on breast, endometrium and cardiovascular health.
Collapse
Affiliation(s)
- Kate Maclaran
- West London Menopause & PMS Centre, Department of Gynaecology, Queen Charlotte's & Chelsea Hospital, Du Cane Road, London W12 0HS, UK
| | | |
Collapse
|
20
|
Fourkala EO, Zaikin A, Burnell M, Gentry-Maharaj A, Ford J, Gunu R, Soromani C, Hasenbrink G, Jacobs I, Dawnay A, Widschwendter M, Lichtenberg-Fraté H, Menon U. Association of serum sex steroid receptor bioactivity and sex steroid hormones with breast cancer risk in postmenopausal women. Endocr Relat Cancer 2012; 19:137-47. [PMID: 22199143 PMCID: PMC3322660 DOI: 10.1530/erc-11-0310] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Postmenopausal women with elevated serum sex steroids have an increased risk of breast cancer. Most of this risk is believed to be exerted through binding of the sex steroids to their receptors. For the first time, we investigate the association of estrogen receptor (ER) and androgen receptor (AR) serum bioactivity (SB) in addition to hormone levels in samples from women with breast cancer collected before diagnosis. Two hundred postmenopausal women participating in the UK Collaborative Trial of Ovarian Cancer Screening who developed ER-positive breast cancer 0.6-5 years after sample donation were identified and matched to 400 controls. ER and AR bioassays were used to measure ERα, ERβ, and AR SB. Androgen and estrogen levels were measured with immunoassays. Subjects were classified according to quintiles of the respective marker among controls and the associations between SB and hormones with breast cancer risk were determined by logistic regression analysis. ERα and ERβ SB were significantly higher before diagnosis compared with controls, while estrogens showed no difference. Women had a twofold increased breast cancer risk if ERα SB (odds ratio (OR), 2.114; 95% confidence interval (CI), 1.050-4.425; P=0.040) was in the top quintile >2 years before diagnosis or estrone (OR, 2.205; 95% CI, 1.104-4.586; P=0.029) was in the top quintile <2 years before diagnosis. AR showed no significant association with breast cancer while androstenedione (OR, 3.187; 95% CI, 1.738-6.044; P=0.0003) and testosterone (OR, 2.145; 95% CI, 1.256-3.712; P=0.006) were significantly higher compared with controls and showed a strong association with an almost threefold increased breast cancer risk independent of time to diagnosis. This study provides further evidence on the association of androgens and estrogens with breast cancer. In addition, it reports that high ER but not AR SB is associated with increased breast risk >2 years before diagnosis.
Collapse
Affiliation(s)
- Evangelia-Ourania Fourkala
- Department of Gynecological Oncology, Institute for Women's Health, Gynecological Cancer Research Centre, University College London, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Wills S, Ravipati A, Venuturumilli P, Kresge C, Folkerd E, Dowsett M, Hayes DF, Decker DA. Effects of vaginal estrogens on serum estradiol levels in postmenopausal breast cancer survivors and women at risk of breast cancer taking an aromatase inhibitor or a selective estrogen receptor modulator. J Oncol Pract 2012; 8:144-8. [PMID: 22942807 DOI: 10.1200/jop.2011.000352] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2011] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Intravaginal estradiols (VE) have been proposed as safe alternatives to systemic estrogen therapy in breast cancer survivors. PATIENTS AND METHODS Postmenopausal women with estrogen receptor-positive breast cancer or at high risk for breast cancer (n = 24) who were taking an aromatase inhibitor (AI) or a selective estrogen receptor modulator (SERM) and VE for ≥ 90 days for atrophic vaginitis and 24 controls taking AI only participated in the study. Serum samples were drawn from VE ring patients before insertion and 30 and 60 days postinsertion, from VE tablet patients the morning before insertion and approximately 12 hours postinsertion, and once from controls. Samples were assayed for E2 concentrations by using highly sensitive radioimmunoassay after ether extraction. RESULTS Mean E2 levels in controls were 3.72 pmol/L (range, < 3.0-7.7 pmol/L); mean E2 levels preinsertion and 12 weeks postinsertion in the VE ring patients were significantly greater than controls (P < .001 for each comparison). Mean preinsertion E2 levels in patients using VE tablets were not significantly different than those of controls (P = .48), and postinsertion levels were 76 pmol/L higher than preinsertion (P < .001). CONCLUSION VE treatment increased E2 levels. Preinsertion levels for patients receiving VE tablets were not elevated compared with those of controls, suggesting that E2 elevations with this preparation may not be continuously sustained. We conclude that VE treatment, regardless of type, results in elevated circulating E2 levels in this population and should be used with caution.
Collapse
Affiliation(s)
- Shannon Wills
- William Beaumont Hospital, Royal Oak; University of Michigan Comprehensive Cancer Center, Ann Arbor, MI; and The Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Akkoc A, Inan S, Sonmez G. Matrix metalloproteinase (MMP-2 and MMP-9) and steroid receptor expressions in feline mammary tumors. Biotech Histochem 2012; 87:312-9. [DOI: 10.3109/10520295.2011.652173] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
23
|
Abstract
The role of androgens on breast cancer development and progression has not been fully elucidated. Several in vivo and in vitro studies demonstrate that androgens have an inhibitory effect on the mammary epithelium, whereas the majority of epidemiological studies report a positive association between high androgen levels and risk of breast cancer. Expression of the androgen receptor is a positive prognostic factor. Understanding the role of androgens in breast carcinogenesis is important because many women use testosterone replacement for the alleviation of symptoms brought on by menopause, in particular high-risk women who undergo surgical menopause at an early age. We overview the literature examining a role of androgens in the etiology of breast cancer.
Collapse
|
24
|
Abstract
Abundant clinical evidence suggests that androgens normally inhibit mammary epithelial proliferation and breast growth. Clinical and nonhuman primate studies support the notion that androgens inhibit mammary proliferation and, thus, may protect from breast cancer. On the other hand, administration of conventional estrogen treatment suppresses endogenous androgens and may, thus, enhance estrogenic breast stimulation and possibly breast cancer risk. Addition of testosterone to the usual hormone therapy regimen may diminish the estrogen/progestin increase in breast cancer risk, but the impact of this combined use on mammary gland homeostasis still needs evaluation.
Collapse
Affiliation(s)
- Constantine Dimitrakakis
- Developmental Endocrinology Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1103, USA.
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW To examine the recent data pertaining to the relationships between testosterone and cardiovascular disease (CVD) and cancer in women. RECENT FINDINGS Despite the entrenched belief that higher blood levels of testosterone increase the risk of CVD in women, data from recent observational studies mostly show an inverse relationship between testosterone and CVD risk. One pilot study suggests favorable effects of nonoral testosterone treatment of women with established congestive cardiac failure which merits further evaluation. The relationship between endogenous testosterone production and breast cancer risk remains contentious, with recent studies indicating either no relationship, or a possible increase in risk when estrone and estradiol are not taken into account. No randomized controlled trial of testosterone therapy has been sufficiently large or of sufficient duration to establish whether such treatment may influence breast cancer occurrence. There does not appear to be an association between testosterone and endometrial cancer, or other malignancies on review of published studies. SUMMARY Testosterone is inversely associated with increased CVD risk in women, whereas low sex hormone binding globulin increases CVD risk. The relationship between testosterone and breast cancer remains unclear, although a clear signal of risk has not emerged from studies of women treated with testosterone therapy over the past decade.
Collapse
Affiliation(s)
- Susan R Davis
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
26
|
Affiliation(s)
- Gerald M Higa
- a Schools of Pharmacy and Medicine, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506-9520, USA.
| |
Collapse
|
27
|
Farhat GN, Cummings SR, Chlebowski RT, Parimi N, Cauley JA, Rohan TE, Huang AJ, Vitolins M, Hubbell FA, Manson JE, Cochrane BB, Lane DS, Lee JS. Sex hormone levels and risks of estrogen receptor-negative and estrogen receptor-positive breast cancers. J Natl Cancer Inst 2011; 103:562-70. [PMID: 21330633 DOI: 10.1093/jnci/djr031] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Endogenous sex hormone levels are associated with risks of breast cancer overall and estrogen receptor (ER)-positive breast tumors; however, their associations with ER-negative tumors remain unclear. METHODS In a case-cohort study within the Women's Health Initiative Observational Study among postmenopausal women aged 50-79 years, we examined associations between endogenous testosterone and estradiol levels and the risks of ER-negative and ER-positive breast cancers. Serum levels of bioavailable testosterone and estradiol were assessed at the baseline visit in 317 invasive breast cancer case subjects and in a subcohort of 594 women. Bioavailable sex hormone levels were calculated using the total hormone level and the sex hormone-binding globulin concentration (measured by radioimmunoassays and a chemiluminescent immunoassay, respectively). Cox proportional hazards regression was used for statistical analysis. All statistical tests were two-sided. RESULT The unadjusted absolute rates of ER-negative breast cancer for testosterone quartiles 1-4 were 0.34, 0.20, 0.23, and 0.21 per 10,000 person-years, respectively. Compared with women in the lowest quartile of testosterone level, those in quartile 2 had a 56% lower risk of ER-negative cancer (hazard ratio [HR] = 0.44, 95% confidence interval [CI] = 0.23 to 0.85), those in quartile 3 had a 45% lower risk (HR = 0.55, 95% CI = 0.30 to 1.01), and those in quartile 4 had a 49% lower risk (HR = 0.51, 95% CI = 0.28 to 0.94), independent of other risk factors. Estradiol level was not associated with ER-negative breast cancer. ER-positive breast cancer risk increased with higher testosterone levels (P(trend) = .04), but this trend was not statistically significant after adjustment for estradiol (P(trend) = .15). ER-positive cancer risk was approximately twofold higher in women with estradiol levels in quartiles 2-4 compared with women in quartile 1, independent of risk factors. CONCLUSION Higher serum levels of bioavailable testosterone are associated with lower risks of ER-negative breast cancer in postmenopausal women.
Collapse
Affiliation(s)
- Ghada N Farhat
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA 94107, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Finch A, Metcalfe KA, Chiang JK, Elit L, McLaughlin J, Springate C, Demsky R, Murphy J, Rosen B, Narod SA. The impact of prophylactic salpingo-oophorectomy on menopausal symptoms and sexual function in women who carry a BRCA mutation. Gynecol Oncol 2011; 121:163-8. [PMID: 21216453 DOI: 10.1016/j.ygyno.2010.12.326] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 12/01/2010] [Accepted: 12/06/2010] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Prophylactic salpingo-oophorectomy is recommended to women who carry a BRCA1 or BRCA2 mutation to reduce the risks of breast, ovarian and fallopian tube cancer. We measured the impact of prophylactic salpingo-oophorectomy on menopausal symptoms and sexual functioning in women with a BRCA mutation. METHODS Women who underwent prophylactic salpingo-oophorectomy between October 1, 2002 and June 26, 2008 for a known BRCA1 or BRCA2 mutation were invited to participate. Participants completed questionnaires before prophylactic surgery and again one year after surgery. Measures of sexual functioning and menopausal symptoms before and after surgery were compared. Satisfaction with the decision to undergo prophylactic salpingo-oophorectomy was evaluated. RESULTS 114 women who underwent prophylactic surgery completed questionnaires before and one year after surgery. Subjects who were premenopausal at the time of surgery (n=75) experienced a significant worsening of vasomotor symptoms (hot flashes, night sweats and sweating) and a decline in sexual functioning (desire, pleasure, discomfort and habit). The increase in vasomotor symptoms and the decline in sexual functioning were mitigated by HRT, but symptoms did not return to pre-surgical levels. HRT decreased vaginal dryness and dyspareunia; however, the decrease in sexual pleasure was not alleviated by HRT. Satisfaction with the decision to undergo prophylactic salpingo-oophorectomy remained high regardless of increased vasomotor symptoms and decreased sexual function. CONCLUSIONS Women who undergo prophylactic salpingo-oophorectomy prior to menopause experience an increase in vasomotor symptoms and a decrease in sexual functioning. These symptoms are improved by HRT, but not to pre-surgical levels.
Collapse
Affiliation(s)
- A Finch
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
LaCroix AZ, Powles T, Osborne CK, Wolter K, Thompson JR, Thompson DD, Allred DC, Armstrong R, Cummings SR, Eastell R, Ensrud KE, Goss P, Lee A, Neven P, Reid DM, Curto M, Vukicevic S. Breast cancer incidence in the randomized PEARL trial of lasofoxifene in postmenopausal osteoporotic women. J Natl Cancer Inst 2010; 102:1706-15. [PMID: 21051656 DOI: 10.1093/jnci/djq415] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Currently available selective estrogen receptor modulators reduce the risk of breast cancer, but they are not widely used. In the Postmenopausal Evaluation and Risk-Reduction with Lasofoxifene (PEARL) trial, lasofoxifene was shown to reduce the risk of estrogen receptor-positive (ER+) breast cancer, nonvertebral and vertebral fractures, coronary artery disease, and stroke, but the effects on total breast cancer (invasive and ductal carcinoma in situ, ER+ and estrogen receptor-negative [ER-]) and ER+ invasive breast cancer are unknown. METHODS Postmenopausal women (n = 8556) aged 59-80 years with low bone density and normal mammograms were randomly assigned to two doses of lasofoxifene (0.25 and 0.5 mg) or placebo. The primary endpoints of the PEARL trial were incidence of ER+ breast cancer and nonvertebral fractures at 5 years. A nested case-control study of 49 incident breast cancer case patients and 156 unaffected control subjects from the PEARL trial was performed to evaluate treatment effects on risk of total and ER+ invasive breast cancer by baseline serum estradiol and sex hormone-binding globulin levels using logistic regression models. Cox proportional hazards models were used to evaluate risk of total breast cancer and ER+ invasive breast cancer using intention-to-treat analysis. All statistical tests were two-sided. RESULTS Breast cancer was confirmed in 49 women. Compared with placebo, 0.5 mg of lasofoxifene statistically significantly reduced the risk of total breast cancer by 79% (hazard ratio = 0.21; 95% confidence interval [CI] = 0.08 to 0.55) and ER+ invasive breast cancer by 83% (hazard ratio = 0.17; 95% CI = 0.05 to 0.57). The effects of 0.5 mg of lasofoxifene on total breast cancer were similar regardless of Gail score, whereas the effects were markedly stronger for women with baseline estradiol levels greater than the median (odds ratio = 0.11; 95% CI = 0.02 to 0.51) vs those with levels less than the median (odds ratio = 0.78; 95% CI = 0.16 to 3.79; P(interaction) = .04). CONCLUSION A 0.5-mg dose of lasofoxifene appears to reduce the risks of both total and ER+ invasive breast cancer in postmenopausal women with osteoporosis.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Anticarcinogenic Agents/therapeutic use
- Biomarkers, Tumor/metabolism
- Bone Density/drug effects
- Bone Density Conservation Agents/therapeutic use
- Breast Neoplasms/epidemiology
- Breast Neoplasms/metabolism
- Breast Neoplasms/prevention & control
- Carcinoma, Ductal, Breast/epidemiology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/prevention & control
- Case-Control Studies
- Double-Blind Method
- Estradiol/blood
- Female
- Fractures, Bone/complications
- Fractures, Bone/etiology
- Fractures, Bone/prevention & control
- Humans
- Incidence
- Mammography
- Middle Aged
- Odds Ratio
- Osteoporosis, Postmenopausal/complications
- Osteoporosis, Postmenopausal/prevention & control
- Primary Prevention/methods
- Pyrrolidines/therapeutic use
- Receptors, Estrogen/blood
- Risk Assessment
- Selective Estrogen Receptor Modulators/therapeutic use
- Sex Hormone-Binding Globulin/metabolism
- Tetrahydronaphthalenes/therapeutic use
- Treatment Outcome
- United States/epidemiology
Collapse
Affiliation(s)
- Andrea Z LaCroix
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Traish AM, Fetten K, Miner M, Hansen ML, Guay A. Testosterone and risk of breast cancer: appraisal of existing evidence. Horm Mol Biol Clin Investig 2010; 2:177-90. [PMID: 25961191 DOI: 10.1515/hmbci.2010.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Accepted: 03/08/2010] [Indexed: 11/15/2022]
Abstract
The objective of this review was to examine data from preclinical, clinical and epidemiological studies to evaluate if testosterone (T) poses increased risk of breast cancer in women. Appraisal of the existing literature produced several lines of evidence arguing against increased breast cancer risk with T. These include: (i) Data from breast tumor cell lines treated with androgens did not corroborate the notion that T increases breast cancer risk. On the contrary, androgens appear to be protective, as they inhibit tumor cell growth. (ii) Many of the epidemiological studies claiming an association between T and breast cancer did not adjust for estrogen levels. Studies adjusted for estrogen levels reported no association between T and breast cancer. (iii) Data from clinical studies with exogenous androgen treatment of women with endocrine and sexual disorders did not show any increase in incidence of breast cancer. (iv) Women afflicted with polycystic ovary disease, who exhibit high levels of androgens do not show increased risk of breast cancer compared to the general population. (v) Female to male transsexuals, who receive supraphysiological doses of T for long time periods prior to surgical procedures, do not report increased risk of breast cancer. (vi) Finally, women with hormone responsive primary breast cancer are treated with aromatase inhibitors, which block conversion of androgens to estrogens, thus elevating androgen levels. These women do not experience increased incidence of contralateral breast cancer nor do they experience increased tumor growth. In conclusion, the evidence available strongly suggests that T does not increase breast cancer risk in women.
Collapse
|
31
|
Vogel VG, Costantino JP, Wickerham DL, Cronin WM, Cecchini RS, Atkins JN, Bevers TB, Fehrenbacher L, Pajon ER, Wade JL, Robidoux A, Margolese RG, James J, Runowicz CD, Ganz PA, Reis SE, McCaskill-Stevens W, Ford LG, Jordan VC, Wolmark N. Update of the National Surgical Adjuvant Breast and Bowel Project Study of Tamoxifen and Raloxifene (STAR) P-2 Trial: Preventing breast cancer. Cancer Prev Res (Phila) 2010; 3:696-706. [PMID: 20404000 DOI: 10.1158/1940-6207.capr-10-0076] [Citation(s) in RCA: 433] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The selective estrogen-receptor modulator (SERM) tamoxifen became the first U.S. Food and Drug Administration (FDA)-approved agent for reducing breast cancer risk but did not gain wide acceptance for prevention, largely because it increased endometrial cancer and thromboembolic events. The FDA approved the SERM raloxifene for breast cancer risk reduction following its demonstrated effectiveness in preventing invasive breast cancer in the Study of Tamoxifen and Raloxifene (STAR). Raloxifene caused less toxicity (versus tamoxifen), including reduced thromboembolic events and endometrial cancer. In this report, we present an updated analysis with an 81-month median follow-up. STAR women were randomly assigned to receive either tamoxifen (20 mg/d) or raloxifene (60 mg/d) for 5 years. The risk ratio (RR; raloxifene:tamoxifen) for invasive breast cancer was 1.24 (95% confidence interval [CI], 1.05-1.47) and for noninvasive disease, 1.22 (95% CI, 0.95-1.59). Compared with initial results, the RRs widened for invasive and narrowed for noninvasive breast cancer. Toxicity RRs (raloxifene:tamoxifen) were 0.55 (95% CI, 0.36-0.83; P = 0.003) for endometrial cancer (this difference was not significant in the initial results), 0.19 (95% CI, 0.12-0.29) for uterine hyperplasia, and 0.75 (95% CI, 0.60-0.93) for thromboembolic events. There were no significant mortality differences. Long-term raloxifene retained 76% of the effectiveness of tamoxifen in preventing invasive disease and grew closer over time to tamoxifen in preventing noninvasive disease, with far less toxicity (e.g., highly significantly less endometrial cancer). These results have important public health implications and clarify that both raloxifene and tamoxifen are good preventive choices for postmenopausal women with elevated risk for breast cancer.
Collapse
|
32
|
Woolcott CG, Shvetsov YB, Stanczyk FZ, Wilkens LR, White KK, Caberto C, Henderson BE, Le Marchand L, Kolonel LN, Goodman MT. Plasma sex hormone concentrations and breast cancer risk in an ethnically diverse population of postmenopausal women: the Multiethnic Cohort Study. Endocr Relat Cancer 2010; 17:125-34. [PMID: 19903744 PMCID: PMC2880171 DOI: 10.1677/erc-09-0211] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
To add to the existing evidence that comes mostly from White populations, we conducted a nested case-control study to examine the association between sex hormones and breast cancer risk within the Multiethnic Cohort that includes Japanese American, White, Native Hawaiian, African American, and Latina women. Of the postmenopausal women for whom we had a plasma sample, 132 developed breast cancer during follow-up. Two controls per case, matched on study area (Hawaii, Los Angeles), ethnicity/race, birth year, date and time of blood draw and time fasting, were randomly selected from the women who had not developed breast cancer. Levels of estradiol (E(2)), estrone (E(1)), androstenedione, dehydroepiandrosterone (DHEA), and testosterone were quantified by RIA after organic extraction and Celite column partition chromatography. E(1) sulfate, DHEA sulfate (DHEAS), and sex hormone-binding globulin (SHBG) were quantified by direct immunoassays. Based on conditional logistic regression, the sex hormones were positively associated and SHBG was negatively associated with breast cancer risk. All associations, except those with DHEAS and testosterone showed a significant linear trend. The odds ratio (OR) associated with a doubling of E(2) levels was 2.26 (95% confidence interval (CI) 1.58-3.25), and the OR associated with a doubling of testosterone levels was 1.34 (95% CI 0.98-1.82). The associations in Japanese American women, who constituted 54% of our sample, were similar to or nonsignificantly stronger than in the overall group. This study provides the best evidence to date that the association between sex hormones and breast cancer risk is generalizable to an ethnically diverse population.
Collapse
Affiliation(s)
- Christy G Woolcott
- Epidemiology Program, Cancer Research Center of Hawaii, University of Hawaii, 1236 Lauhala Street, Honolulu, Hawaii 96813, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Davis SR, Hirschberg AL, Wagner LK, Lodhi I, von Schoultz B. The effect of transdermal testosterone on mammographic density in postmenopausal women not receiving systemic estrogen therapy. J Clin Endocrinol Metab 2009; 94:4907-13. [PMID: 19850682 DOI: 10.1210/jc.2009-1523] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
CONTEXT Greater mammographic density is associated with increased breast cancer risk and reduced diagnostic mammographic sensitivity and may be seen with estrogen/progestin therapy (EPT). The effects of testosterone therapy on mammographic density in postmenopausal women not on EPT are not known. OBJECTIVE Our objective was to compare effects of two doses of the testosterone transdermal patch (TTP) with placebo in postmenopausal women without concomitant EPT on mammographic density over 52 wk. DESIGN We conducted a randomized, double-blind, placebo-controlled, parallel-group, multinational trial. PATIENTS Patients included 279 postmenopausal women participating in a testosterone and sexual function study with paired mammograms for baseline and 52 wk/exit. INTERVENTIONS Patients were randomized to placebo, TTP 150 microg/d, or TTP 300 microg/d, stratified by menopause type (natural or surgical). MAIN OUTCOME MEASURES Change from baseline to wk 52 in the percentage of dense tissue (PD) on digital mammograms. RESULTS A total of 250 women with paired mammograms for study baseline and wk 52 were included in the primary analysis. Mean age was 54.6 yr, baseline body mass index was 27.5 kg/m(2), and 78% were naturally menopausal. There were no baseline differences between groups. Mean changes from baseline (+/-SEM) in PD for placebo, TTP 150 microg/d and TTP 300 microg/d were small (0.05 +/- 0.16, 0.06 +/- 0.19, and 0.21 +/- 0.17%) and not significantly different. There were no statistically significant differences from placebo for total dense or nondense area and no significant relationships between hormone levels and PD after adjustment for body mass index. CONCLUSION TTP therapy over 52 wk appears to have no significant effect on digitally quantified absolute or percent dense mammographic area in postmenopausal women not using EPT.
Collapse
Affiliation(s)
- Susan R Davis
- Women's Health Program, Department of Medicine, Central and Eastern Clinical School, Monash University, Alfred Hospital, Commercial Road, Prahran, Victoria 3181, Australia.
| | | | | | | | | |
Collapse
|
34
|
Mousa NA, Jebrail MJ, Yang H, Abdelgawad M, Metalnikov P, Chen J, Wheeler AR, Casper RF. Droplet-Scale Estrogen Assays in Breast Tissue, Blood, and Serum. Sci Transl Med 2009; 1:1ra2. [DOI: 10.1126/scitranslmed.3000105] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
35
|
Wood CE, Lees CJ, Cline JM. Mammary gland and endometrial effects of testosterone in combination with oral estradiol and progesterone. Menopause 2009; 16:466-76. [PMID: 19265727 PMCID: PMC2755604 DOI: 10.1097/gme.0b013e318191747a] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The goal of this pilot study was to evaluate the effects of testosterone (T) cotherapy on mammary gland and endometrial measures in a postmenopausal primate model. METHODS Twenty-five surgically postmenopausal cynomolgus monkeys were randomized by social group to receive daily treatment with (1) placebo, (2) oral micronized 17beta-estradiol (1 mg/d equivalent in women) + progesterone (200 mg/d equivalent in women) (E + P), or (3) E + P with T administered via subcutaneous pellets for 8 weeks at a high dose (15 mg) followed by 8 weeks at a low dose (1.5 mg) (E + P + T). The main outcome measures were breast and endometrial epithelial proliferation, as measured by Ki67/MIB1 immunolabeling. RESULTS Intralobular breast proliferation did not differ significantly among groups after 8 weeks of treatment but was marginally higher (P = 0.03) in the E + P + T group after 16 weeks of treatment. No significant increase in proliferation was seen for E + P alone. Comparable changes in mammary gland markers of estrogen-receptor activity were seen for the E + P and E + P + T groups. In the endometrium, the addition of T did not increase endometrial glandular proliferation or estrogen-receptor activity or result in any distinct histologic changes. CONCLUSIONS The findings of this study do not support the idea that T antagonizes the effects of combined hormone therapy on breast proliferation or markers of estrogen-receptor activity. Overall, the short-term effects of T cotherapy on the mammary gland and endometrium were minimal.
Collapse
Affiliation(s)
- Charles E Wood
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA.
| | | | | |
Collapse
|
36
|
Shufelt CL, Braunstein GD. Safety of testosterone use in women. Maturitas 2009; 63:63-6. [DOI: 10.1016/j.maturitas.2009.01.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Accepted: 01/20/2009] [Indexed: 10/21/2022]
|
37
|
Wang B, Mi M, Wang J, Wei N, Zhang Q, Zhu J, Yang S, Guo B, Xu J, Yang X. Does the increase of endogenous steroid hormone levels also affect breast cancer risk in Chinese women? A case-control study in Chongqing, China. Int J Cancer 2009; 124:1892-9. [PMID: 19117053 DOI: 10.1002/ijc.24132] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Accumulating epidemiological evidence suggests that sex steroid hormones are positively associated with the development of breast cancer. However, most of these studies were conducted among Caucasian women and few have been carried out in China. To determine whether the associations of sex steroid hormone levels with breast cancer risk observed by and large in Caucasian populations are also evident in Chinese women, we conducted a case-control study in Chongqing, China. The study included 367 incident breast cancer patients and 367 healthy controls matched on menstrual status, age and periods of blood collection in the menstrual cycle. Plasma concentrations of estradiol, progesterone, testosterone, dehydroepiandrosterone sulfate (DHEAS) and sex hormone binding globulin (SHBG) were determined by electrochemiluminescene immunoassay (ECLIA). Conditional logistic regression analysis was performed to examine their associations with breast cancer risk. From comparisons of upper and lower tertiles, we observed statistically significant positive associations with breast cancer risk for plasma estradiol levels in follicular phase (adjusted odds ratio [OR] = 5.48, 95% confidence interval [CI] = 1.58-18.97), luteal phase (OR = 4.23, CI = 1.65-10.87) and postmenopausal (OR = 2.67, CI = 1.20-5.93); for progesterone levels in luteal phase (OR = 3.11, CI = 1.28-7.56), and for testosterone levels in postmenopausal (OR = 2.83, CI = 1.26-6.35). No significant association was found with DHEAS or SHBG. Our study suggests that high circulating levels of estradiol and testosterone are positively associated with increased breast cancer risk in Chinese women, which are generally consistent with the observations in Caucasian populations.
Collapse
Affiliation(s)
- Bin Wang
- Department of Nutrition and Food Hygiene, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Cummings SR, Tice JA, Bauer S, Browner WS, Cuzick J, Ziv E, Vogel V, Shepherd J, Vachon C, Smith-Bindman R, Kerlikowske K. Prevention of breast cancer in postmenopausal women: approaches to estimating and reducing risk. J Natl Cancer Inst 2009; 101:384-98. [PMID: 19276457 DOI: 10.1093/jnci/djp018] [Citation(s) in RCA: 180] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND It is uncertain whether evidence supports routinely estimating a postmenopausal woman's risk of breast cancer and intervening to reduce risk. METHODS We systematically reviewed prospective studies about models and sex hormone levels to assess breast cancer risk and used meta-analysis with random effects models to summarize the predictive accuracy of breast density. We also reviewed prospective studies of the effects of exercise, weight management, healthy diet, moderate alcohol consumption, and fruit and vegetable intake on breast cancer risk, and used random effects models for a meta-analyses of tamoxifen and raloxifene for primary prevention of breast cancer. All studies reviewed were published before June 2008, and all statistical tests were two-sided. RESULTS Risk models that are based on demographic characteristics and medical history had modest discriminatory accuracy for estimating breast cancer risk (c-statistics range = 0.58-0.63). Breast density was strongly associated with breast cancer (relative risk [RR] = 4.03, 95% confidence interval [CI] = 3.10 to 5.26, for Breast Imaging Reporting and Data System category IV vs category I; RR = 4.20, 95% CI = 3.61 to 4.89, for >75% vs <5% of dense area), and adding breast density to models improved discriminatory accuracy (c-statistics range = 0.63-0.66). Estradiol was also associated with breast cancer (RR range = 2.0-2.9, comparing the highest vs lowest quintile of estradiol, P < .01). Most studies found that exercise, weight reduction, low-fat diet, and reduced alcohol intake were associated with a decreased risk of breast cancer. Tamoxifen and raloxifene reduced the risk of estrogen receptor-positive invasive breast cancer and invasive breast cancer overall. CONCLUSIONS Evidence from this study supports screening for breast cancer risk in all postmenopausal women by use of risk factors and breast density and considering chemoprevention for those found to be at high risk. Several lifestyle changes with the potential to prevent breast cancer should be recommended regardless of risk.
Collapse
Affiliation(s)
- Steven R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, 94107, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Neilson HK, Friedenreich CM, Brockton NT, Millikan RC. Physical activity and postmenopausal breast cancer: proposed biologic mechanisms and areas for future research. Cancer Epidemiol Biomarkers Prev 2009; 18:11-27. [PMID: 19124476 DOI: 10.1158/1055-9965.epi-08-0756] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Convincing evidence now supports a probable preventive role for physical activity in postmenopausal breast cancer. The mechanisms by which long-term physical activity affect risk, however, remain unclear. The aims of this review were to propose a biological model whereby long-term physical activity lowers postmenopausal breast cancer risk and to highlight gaps in the epidemiologic literature. To address the second aim, we summarized epidemiologic literature on 10 proposed biomarkers, namely, body mass index (BMI), estrogens, androgens, sex hormone binding globulin, leptin, adiponectin, markers of insulin resistance, tumor necrosis factor-alpha, interleukin-6, and C-reactive protein, in relation to postmenopausal breast cancer risk and physical activity, respectively. Associations were deemed "convincing," "probable," "possible," or "hypothesized" using set criteria. Our proposed biological model illustrated the co-occurrence of overweight/obesity, insulin resistance, and chronic inflammation influencing cancer risk through interrelated mechanisms. The most convincing epidemiologic evidence supported associations between postmenopausal breast cancer risk and BMI, estrogens, and androgens, respectively. In relation to physical activity, associations were most convincing for BMI, estrone, insulin resistance, and C-reactive protein. Only BMI and estrone were convincingly (or probably) associated with both postmenopausal breast cancer risk and physical activity. There is a need for prospective cohort studies relating the proposed biomarkers to cancer risk and for long-term exercise randomized controlled trials comparing biomarker changes over time, specifically in postmenopausal women. Future etiologic studies should consider interactions among biomarkers, whereas exercise trials should explore exercise effects independently of weight loss, different exercise prescriptions, and effects on central adiposity.
Collapse
Affiliation(s)
- Heather K Neilson
- Division of Population Health, Alberta Cancer Board, 1331-29 Street NW, Calgary, Alberta, Canada T2N 4N2
| | | | | | | |
Collapse
|
40
|
Abstract
Androgens have important physiological effects in women while at the same time they may be implicated in breast cancer pathologies. However, data on the effects of androgens on mammary epithelial proliferation and/or breast cancer incidence are not in full agreement. We performed a literature review evaluating current clinical, genetic and epidemiological data regarding the role of androgens in mammary growth and neoplasia. Epidemiological studies appear to have significant methodological limitations and thus provide inconclusive results. The study of molecular defects involving androgenic pathways in breast cancer is still in its infancy. Clinical and nonhuman primate studies suggest that androgens inhibit mammary epithelial proliferation and breast growth while conventional estrogen treatment suppresses endogenous androgens. Abundant clinical evidence suggests that androgens normally inhibit mammary epithelial proliferation and breast growth. Suppression of androgens using conventional estrogen treatment may thus enhance estrogenic breast stimulation and possibly breast cancer risk. Addition of testosterone to the usual hormone therapy regimen may diminish the estrogen/progestin increase in breast cancer risk but the impact of this combined use on mammary gland homeostasis still needs evaluation.
Collapse
Affiliation(s)
- Constantine Dimitrakakis
- Developmental Endocrinology Branch, National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive, MSC-1103 Bethesda, Maryland 20892-1103, USA
- 1st Department of Ob/Gyn, Athens University Medical School, 80 Vas. Sophias Street, 11528, Athens, Greece
| | - Carolyn Bondy
- Developmental Endocrinology Branch, National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive, MSC-1103 Bethesda, Maryland 20892-1103, USA
| |
Collapse
|
41
|
Abstract
Breast cancer risk factors have been studied for the past three decades, and the single most important risk factor is age. Hormonally linked adult reproductive and anthropometric risk factors contribute to the etiology of postmenopausal breast cancer. The risk of breast cancer increases among women older than 50 years of age who have benign breast disease, especially those with atypical ductal or lobular hyperplasia. Lobular carcinoma in situ increases risk significantly, as do a family history of breast cancer in first-degree relatives and the presence of BRCA1 or BRCA2 mutations. Diet, exercise, and environmental factors play a very small role in overall risk. Mammographic breast density increases relative risk fivefold among women with the highest density, and breast cancer risk is two to three times greater in women with elevated serum levels of estradiol or testosterone. Multivariate risk models allow determination of composite relative risks and cumulative lifetime risk, although improved models for African American women are required. For postmenopausal women, newer risk models are being developed and validated that include age, breast density, race, ethnicity, family history of breast cancer, a previous breast biopsy, body mass index, age at onset of natural menopause, hormone therapy, and previous false-positive mammography. A simpler model that includes only age, breast cancer in first-degree relatives, and previous breast biopsy performs well for estrogen receptor-positive breast cancer in postmenopausal women. As many as 10 million women in the United States are at increased risk, and clinicians are obligated to identify these women and manage their risk appropriately.
Collapse
|
42
|
Meyer zu Schwabedissen HE, Tirona RG, Yip CS, Ho RH, Kim RB. Interplay between the nuclear receptor pregnane X receptor and the uptake transporter organic anion transporter polypeptide 1A2 selectively enhances estrogen effects in breast cancer. Cancer Res 2008; 68:9338-47. [PMID: 19010908 DOI: 10.1158/0008-5472.can-08-0265] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The ligand-activated nuclear receptor pregnane X receptor (PXR) is known to play a role in the regulated expression of drug metabolizing enzymes and transporters. Recent studies suggest a potential clinically relevant role of PXR in breast cancer. However, the relevant pathway or target genes of PXR in breast cancer biology and progression have not yet been fully clarified. In this study, we show that mRNA expression of organic anion transporter polypeptide 1A2 (OATP1A2), a transporter capable of mediating the cellular uptake of estrogen metabolites, is nearly 10-fold greater in breast cancer compared with adjacent healthy breast tissues. Immunohistochemistry revealed exclusive expression of OATP1A2 in breast cancer tissue. Interestingly, treatment of breast cancer cells in vitro with the PXR agonist rifampin induced OATP1A2 expression in a time-dependent and concentration-dependent manner. Consistent with its role as a hormone uptake transporter, induction of OATP1A2 was associated with increased uptake of estrone 3-sulfate. The rifampin response was abrogated after small interfering RNA targeting of PXR. We then identified a PXR response element in the human OATP1A2 promoter, located approximately 5.7 kb upstream of the transcription initiation site. The specificity of PXR-OATP1A2 promoter interaction was confirmed using chromatin immunoprecipitation. Importantly, we used a novel potent and specific antagonist of PXR (A-792611) to show the reversal of the rifampin effect on the cellular uptake of E(1)S. These data provide important new insights into the interplay between a xenobiotic nuclear receptor PXR and OATP1A2 that could contribute to the pathogenesis of breast cancer and may also prove to be heretofore unrecognized targets for breast cancer treatment.
Collapse
|
43
|
Abstract
Although women have been treated with testosterone (T) for female sexual dysfunction since the 1950s, the role of T in normal female physiology is not yet fully defined. One of the major safety concerns of androgen therapy is whether androgens have a stimulatory effect on the breast that could lead to breast carcinomas. The proposed mechanisms for such stimulation include local estrogen production from the aromatase enzyme complex present in the breast tissue or by the direct stimulation of the androgen receptor. Predominant data from in vitro studies have shown that androgens actually have apoptotic and antiproliferative effects and not stimulatory effects. Animal models have shown similar results to in vitro studies, finding that androgens inhibit breast cancer growth. Prospective and retrospective epidemiological analyses have shown mixed outcomes, with no clear consensus regarding androgen use and breast cancer risk. Hyperandrogenism in patients with polycystic ovarian syndrome with elevated levels of endogenous T is not associated with an increased risk of breast cancer and may, in fact, be protective. Another human model with excess of T is female-to-male transgenderism, in which genotypic women are treated with large doses of exogenous T with no increased risk. High-dose androgen therapy also has been effective in treating patients with advanced breast cancer. Thus, the preponderance of data suggests that T use in females is not associated with an increased risk of breast carcinoma.
Collapse
|
44
|
Braunstein GD, Johnson BD, Stanczyk FZ, Bittner V, Berga SL, Shaw L, Hodgson TK, Paul-Labrador M, Azziz R, Merz CNB. Relations between endogenous androgens and estrogens in postmenopausal women with suspected ischemic heart disease. J Clin Endocrinol Metab 2008; 93:4268-75. [PMID: 18728170 PMCID: PMC2582570 DOI: 10.1210/jc.2008-0792] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2008] [Accepted: 08/15/2008] [Indexed: 01/09/2023]
Abstract
CONTEXT Because androgens are obligatory precursors of estrogens, it is reasonable to assume that their serum concentrations would exhibit positive correlations. If so, then epidemiologic studies that examine the association between androgens and pathological processes should adjust the results for the independent effect of estrogens. OBJECTIVE The objective of the study was to examine the interrelationships among testosterone (T), androstenedione, estradiol (E2), estrone, and SHBG in postmenopausal women. DESIGN This was a cross-sectional study of women participating in the National Heart, Blood, and Lung Institute-sponsored Women's Ischemia Syndrome Evaluation study. SETTING The study was conducted at four academic medical centers. PATIENTS A total of 284 postmenopausal women with chest pain symptoms or suspected myocardial ischemia. MAIN OUTCOME MEASURES Post hoc analysis of the relationships among sex steroid hormones with insulin resistance, body mass index (BMI), and presence or absence of coronary artery disease as determined by coronary angiography. RESULTS BMI was significantly associated with insulin resistance, total E2, free E2, bioavailable E2, and free T. Highly significant correlations were found for total T, free T, and androstenedione with total E2, free E2, bioavailable E2, and estrone and persisted after adjustment for BMI and insulin resistance. A significant relationship was present between total and free T and the presence of coronary artery disease after adjustment for the effect of E2. CONCLUSIONS Serum levels of androgens and estrogens track closely in postmenopausal women referred for coronary angiography for suspected myocardial ischemia. Epidemiological studies that relate sex steroid hormones to physiological or pathological processes need to control for the independent effect of both estrogens and androgens.
Collapse
Affiliation(s)
- Glenn D Braunstein
- Department of Medicine, Room 2119, Plaza Level, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, California 90048, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Papageorge MB. Etiology of oral cancer in the young patient: is tongue cancer becoming the other cancer in women? Oral Maxillofac Surg Clin North Am 2008; 19:163-71, v. [PMID: 18088875 DOI: 10.1016/j.coms.2007.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The recently reported increase in the incidence of oral cancer in a younger population, especially in young women, in the absence of the traditional risk factors has raised concern. Of particular interest is the increased incidence of carcinoma of the tongue, which has been suggested to be a distinct entity. Although there are conflicting data on incidence and sex distribution, studies suggest that the physiologic response to risk factors by men and women and the clinical behavior of these cancers in the younger population may be different than the normal variant. Effort is currently being made to elucidate the etiology and pathogenesis of oral cancer in the younger population.
Collapse
Affiliation(s)
- Maria B Papageorge
- Department of Oral and Maxillofacial Surgery, Tufts University School of Dental Medicine, One Kneeland Street, Boston, MA 02111, USA.
| |
Collapse
|
46
|
Abstract
Exposure to higher levels of both exogenous and endogenous hormone is associated with breast cancer risk. Because of the association between breast cancer and HRT, only the minimal duration of HRT use is recommended for symptom control, and it is not recommended for chronic disease management. Current research issues include the role of progestins, other types of HRT, duration of unopposed estrogen use, and characteristics of cancers that develop on HRT. Circulating sex steroid levels are associated with breast cancer risk, but multiple issues need to be addressed before they are used routinely in clinical practice. Current research issues include measurement of levels for routine clinical practice, integration with standard breast cancer risk models and genetic polymorphism data, and applicability to estrogen-receptor-negative cancers.
Collapse
Affiliation(s)
- Wendy Y Chen
- Channing Laboratory, Brigham and Women's Hospital, 181 Longwood Avenue, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
47
|
Testosterone, Other Androgens and Breast Cancer. Breast Cancer 2008. [DOI: 10.3109/9781420058734-15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
48
|
Rock CL, Flatt SW, Laughlin GA, Gold EB, Thomson CA, Natarajan L, Jones LA, Caan BJ, Stefanick ML, Hajek RA, Al-Delaimy WK, Stanczyk FZ, Pierce JP. Reproductive steroid hormones and recurrence-free survival in women with a history of breast cancer. Cancer Epidemiol Biomarkers Prev 2008; 17:614-20. [PMID: 18323413 PMCID: PMC2575111 DOI: 10.1158/1055-9965.epi-07-0761] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Epidemiologic studies fairly consistently show in postmenopausal women that reproductive steroid hormones contribute to primary breast cancer risk, and this association is strongly supported by experimental studies using laboratory animals and model systems. Evidence linking sex hormone concentrations with risk for recurrence in women diagnosed with breast cancer is limited; however, beneficial effects of antiestrogenic therapy on recurrence-free survival suggest that these hormones affect progression and risk for recurrence. This study examined whether baseline serum concentrations of estradiol, testosterone, and sex hormone binding globulin were associated with recurrence-free survival in a nested case-control cohort of women from a randomized diet trial (Women's Healthy Eating and Living Study) who were followed for >7 years after diagnosis. In 153 case-control pairs of perimenopausal and postmenopausal women in this analysis, total estradiol [hazard ratio (HR), 1.41 per unit increase in log concentration; 95% confidence interval (95% CI), 1.01-1.97], bioavailable estradiol (HR, 1.26; 95% CI, 1.03-1.53), and free estradiol (HR, 1.31; 95% CI, 1.03-1.65) concentrations were significantly associated with risk for recurrence. Recurred women had an average total estradiol concentration that was double that of nonrecurred women (22.7 versus 10.8 pg/mL; P = 0.05). Testosterone and sex hormone binding globulin concentrations did not differ between cases and controls and were not associated with risk for recurrence. Although genetic and metabolic factors likely modulate the relationship between circulating sex hormones and risk, results from this study provide evidence that higher serum estrogen concentration contributes to risk for recurrence in women diagnosed with early stage breast cancer.
Collapse
Affiliation(s)
- Cheryl L Rock
- Cancer Prevention and Control Program, Moores UCSD Cancer Center, University of California-San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0901, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Endogenous Hormone Levels and Risk of Breast, Endometrial and Ovarian Cancers:. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008. [DOI: 10.1007/978-0-387-78818-0_10] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
50
|
Chlebowski RT, Anderson GL, Lane DS, Aragaki AK, Rohan T, Yasmeen S, Sarto G, Rosenberg CA, Hubbell FA. Predicting risk of breast cancer in postmenopausal women by hormone receptor status. J Natl Cancer Inst 2007; 99:1695-705. [PMID: 18000216 DOI: 10.1093/jnci/djm224] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Strategies for estrogen receptor (ER)-positive breast cancer risk reduction in postmenopausal women require screening of large populations to identify those with potential benefit. We evaluated and attempted to improve the performance of the Breast Cancer Risk Assessment Tool (i.e., the Gail model) for estimating invasive breast cancer risk by receptor status in postmenopausal women. METHODS In The Women's Health Initiative cohort, breast cancer risk estimates from the Gail model and models incorporating additional or fewer risk factors and 5-year incidence of ER-positive and ER-negative invasive breast cancers were determined and compared by use of receiver operating characteristics and area under the curve (AUC) statistics. All statistical tests were two-sided. RESULTS Among 147,916 eligible women, 3236 were diagnosed with invasive breast cancer. The overall AUC for the Gail model was 0.58 (95% confidence interval [CI]=0.56 to 0.60). The Gail model underestimated 5-year invasive breast cancer incidence by approximately 20% (P<.001), mostly among those with a low estimated risk. Discriminatory performance was better for the risk of ER-positive cancer (AUC = 0.60, 95% CI = 0.58 to 0.62) than for the risk of ER-negative cancer (AUC = 0.50, 95% CI = 0.45 to 0.54). Age and age at menopause were statistically significantly associated with ER-positive but not ER-negative cancers (P=.05 and P=.04 for heterogeneity, respectively). For ER-positive cancers, no additional risk factors substantially improved the Gail model prediction. However, a simpler model that included only age, breast cancer in first-degree relatives, and previous breast biopsy examination performed similarly for ER-positive breast cancer prediction (AUC=0.58, 95% CI= 0.56 to 0.60); postmenopausal women who were 55 years or older with either a previous breast biopsy examination or a family history of breast cancer had a 5-year breast cancer risk of 1.8% or higher. CONCLUSIONS In postmenopausal women, the Gail model identified populations at increased risk for ER-positive but not ER-negative breast cancers. A model with fewer variables appears to provide a simpler approach for screening for breast cancer risk.
Collapse
Affiliation(s)
- Rowan T Chlebowski
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, 1124 West Carson Street, Torrance, CA 90502, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|