1
|
Hugh JC, Haddon LSJ, Githaka JM. DREAM On, DREAM Off: A Review of the Estrogen Paradox in Luminal A Breast Cancers. Biomedicines 2024; 12:1300. [PMID: 38927507 PMCID: PMC11201522 DOI: 10.3390/biomedicines12061300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/27/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
It is generally assumed that all estrogen-receptor-positive (ER+) breast cancers proliferate in response to estrogen and, therefore, examples of the estrogen-induced regression of ER+ cancers are paradoxical. This review re-examines the estrogen regression paradox for the Luminal A subtype of ER+ breast cancers. The proliferative response to estrogen is shown to depend on the level of ER. Mechanistically, a window of opportunity study of pre-operative estradiol suggested that with higher levels of ER, estradiol could activate the DREAM-MMB (Dimerization partner, Retinoblastoma-like proteins, E2F4, and MuvB-MYB-MuvB) pathway to decrease proliferation. The response of breast epithelium and the incidence of breast cancers during hormonal variations that occur during the menstrual cycle and at the menopausal transition, respectively, suggest that a single hormone, either estrogen, progesterone or androgen, could activate the DREAM pathway, leading to reversible cell cycle arrest. Conversely, the presence of two hormones could switch the DREAM-MMB complex to a pro-proliferative pathway. Using publicly available data, we examine the gene expression changes after aromatase inhibitors and ICI 182,780 to provide support for the hypothesis. This review suggests that it might be possible to integrate all current hormonal therapies for Luminal A tumors within a single theoretical schema.
Collapse
Affiliation(s)
- Judith C. Hugh
- Department of Laboratory Medicine and Pathology, University of Alberta, 116 St & 85 Ave, Edmonton, AB T6G 2R3, Canada
| | - Lacey S. J. Haddon
- Department of Chemistry, University of Alberta, 116 St & 85 Ave, Edmonton, AB T6G 2R3, Canada;
| | - John Maringa Githaka
- Department of Biochemistry, University of Alberta, 116 St & 85 Ave, Edmonton, AB T6G 2R3, Canada;
| |
Collapse
|
2
|
Wang F, Eikeland E, Reidunsdatter RJ, Hagen L, Engstrøm MJ, Geisler J, Haanpää M, Hämäläinen E, Giskeødegård GF, Bathen TF. Quantification of multiple steroid hormones in serum and human breast cancer tissue by liquid chromatography-tandem mass spectrometry analysis. Front Oncol 2024; 14:1383104. [PMID: 38863629 PMCID: PMC11165045 DOI: 10.3389/fonc.2024.1383104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/06/2024] [Indexed: 06/13/2024] Open
Abstract
Introduction Systemic and local steroid hormone levels may function as novel prognostic and predictive biomarkers in breast cancer patients. We aimed at developing a novel liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for the simultaneous measurement of multiple, biologically pivotal steroid hormones in human serum and breast cancer tissue. Methods The quantitative method consisted of liquid-liquid extraction, Sephadex LH-20 chromatography for tissue extracts, and analysis of steroid hormones by liquid-chromatography-tandem mass spectrometry. We analyzed serum and tissue steroid hormone levels in 16 and 40 breast cancer patients, respectively, and assessed their correlations with clinical parameters. Results The method included quantification of nine steroid hormones in serum [including cortisol, cortisone, corticosterone, estrone (E1), 17β-estradiol (E2), 17α-hydroxyprogesterone, androstenedione (A4), testosterone and progesterone) and six (including cortisone, corticosterone, E1, E2, A4, and testosterone) in cancer tissue. The lower limits of quantification were between 0.003-10 ng/ml for serum (250 µl) and 0.038-125 pg/mg for tissue (20 mg), respectively. Accuracy was between 98%-126%, intra-assay coefficient of variations (CV) was below 15%, and inter-assay CV were below 11%. The analytical recoveries for tissue were between 76%-110%. Tissue levels of E1 were positively correlated with tissue E2 levels (p<0.001), and with serum levels of E1, E2 and A4 (p<0.01). Tissue E2 levels were positively associated with serum E1 levels (p=0.02), but not with serum E2 levels (p=0.12). The levels of tissue E2 and ratios of E1 to A4 levels (an index for aromatase activity) were significantly higher in patients with larger tumors (p=0.03 and p=0.02, respectively). Conclusions The method was convenient and suitable for a specific and accurate profiling of clinically important steroid hormones in serum. However, the sensitivity of the profile method in steroid analysis in tissue samples is limited, but it can be used for the analysis of steroids in breast cancer tissues if the size of the sample or its steroid content is sufficient.
Collapse
Affiliation(s)
- Feng Wang
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Breast and Endocrine of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Eline Eikeland
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Randi J. Reidunsdatter
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Lars Hagen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim, Norway
- PROMEC Core Facility for Proteomics and Modomics, Norwegian University of Science and Technology, and the Central Norway Regional Health Authority Norway, Trondheim, Norway
| | - Monica J. Engstrøm
- Department of Breast and Endocrine of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway & Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Mikko Haanpää
- HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Esa Hämäläinen
- Department of Clinical Chemistry, University of Eastern Finland, Kuopio, Finland
| | - Guro F. Giskeødegård
- Department of Breast and Endocrine of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Tone F. Bathen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
3
|
Honma N, Arai T, Matsuda Y, Fukunaga Y, Akishima-Fukasawa Y, Yamamoto N, Kawachi H, Ishikawa Y, Takeuchi K, Mikami T. Estrogen concentration and estrogen receptor-β expression in postmenopausal colon cancer considering patient/tumor background. J Cancer Res Clin Oncol 2022; 148:1063-1071. [PMID: 35032217 DOI: 10.1007/s00432-021-03889-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 12/13/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE A large number of studies have suggested an inhibitory role of estrogens against colorectal cancer (CRC), but persistent controversy exists. CRC characteristics are affected by sex, age, and tumor locus, suggesting the need for a systematic study considering these factors. The purpose of this study was to verify the difference in the pathobiological role of estrogens in CRC according to patient/tumor backgrounds. METHODS Surgical specimens from 116 postmenopausal women (≥ 70 years/o, n = 74; < 70 years/o, n = 42) were studied. Estrogen receptor-β (ER-β), the main ER in the colorectal epithelium, was immunohistochemically examined. The concentrations of estradiol (E2) and estrone (E1) were examined by liquid chromatography-tandem mass spectrometry (LC-MS/MS). These factors were compared according to the tissue type (cancerous or non-cancerous), patients' age, tumor backgrounds (locus, histology, pathological stage, status of mismatch repair protein = MMR), and clinical outcome. RESULTS ER-β-positivity, higher E2 concentration, deficient-MMR, and medullary/mucinous histology (Med/Muc) were closely related to right-sided tumors in women who were aged ≥ 70 years /o (R-Ca ≥ 70) and also closely related to each other. ER-β reduction compared with non-cancerous counterparts was observed only in left-sided tumors of patients < 70 years /o (L-Ca < 70), non-Med/Muc, or proficient-MMR tumors. CONCLUSION The present results suggest that estrogens do not suppress, but rather promote, R-Ca ≥ 70, Med/Muc, or deficient-MMR tumors, whereas estrogens suppress L-Ca < 70, non-Med/Muc, or proficient-MMR tumors, confirming the difference in pathobiological role of estrogens in postmenopausal colon cancer according to the patients' age and tumor background. This may at least partly explain the controversy regarding the association between estrogens and CRC.
Collapse
Affiliation(s)
- Naoko Honma
- Department of Pathology, Faculty of Medicine, Toho University, Omori-Nishi 5-21-16, Ota-ku, Tokyo, 143-8540, Japan. .,Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan.
| | - Tomio Arai
- Department of Pathology, Tokyo Metropolitan Geriatric Hospital, Sakaecho 35-2, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Yoko Matsuda
- Oncology Pathology, Department of Pathology and Host-Defense, Faculty of Medicine, Kagawa University, Ikenobe 1750-1, Kita-gun, Miki, Kagawa, 761-0793, Japan
| | - Yosuke Fukunaga
- Department of Gastroenterological Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan
| | - Yuri Akishima-Fukasawa
- Department of Pathology, Faculty of Medicine, Toho University, Omori-Nishi 5-21-16, Ota-ku, Tokyo, 143-8540, Japan
| | - Noriko Yamamoto
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan.,Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan
| | - Hiroshi Kawachi
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan.,Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan
| | - Yuichi Ishikawa
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan.,Department of Pathology, International University of Health and Welfare, Mita 1-4-3, Minato-ku, Tokyo, 108-8329, Japan
| | - Kengo Takeuchi
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan.,Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Ariake 3-8-31, Koto-ku, Tokyo, 135-8550, Japan
| | - Tetuo Mikami
- Department of Pathology, Faculty of Medicine, Toho University, Omori-Nishi 5-21-16, Ota-ku, Tokyo, 143-8540, Japan
| |
Collapse
|
4
|
Caceres S, Monsalve B, Alonso-Diez A, Crespo B, Illera MJ, de Andres PJ, Silvan G, Illera JC. Blocking Estrogen Synthesis Leads to Different Hormonal Responses in Canine and Human Triple Negative Inflammatory Breast Cancer. Cancers (Basel) 2021; 13:cancers13194967. [PMID: 34638451 PMCID: PMC8507680 DOI: 10.3390/cancers13194967] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 11/16/2022] Open
Abstract
Blocking estrogen synthesis by inhibitors of estrogen synthesis is a widely used therapy against estrogen receptor-positive tumors. However, these therapies are less effective in negative expression tumors. Therefore, this study determined the effectiveness of anti-aromatase and anti-sulfatase therapies in canine and human inflammatory breast cancer. Cell cultures and xenografts from IPC-366 and SUM149 were treated with different doses of letrozole (anti-aromatase) and STX-64 (anti-sulfatase), in order to observe their effectiveness in terms of cell proliferation, tumor progression, and the appearance of metastases and hormonal profiles. The results revealed that both treatments are effective in vitro since they reduce cell proliferation and decrease the secreted estrogen levels. In xenograft mice, while treatment with letrozole reduces tumor progression by 30-40%, STX-64 increases tumor progression by 20%. The hormonal results obtained determined that STX-64 produced an increase in circulating and intratumoral levels of estradiol, which led to an increase in tumor progression. However, letrozole was able to block estrogen synthesis by decreasing the levels of circulating and intratumoral estrogen and thus slowing down tumor progression. In conclusion, letrozole can be an effective treatment for canine and human inflammatory breast cancer. The knowledge of the hormonal profile of breast tumors reflects useful information on the effectiveness of different endocrine treatments.
Collapse
Affiliation(s)
- Sara Caceres
- Department of Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (B.M.); (B.C.); (M.J.I.); (G.S.); (J.C.I.)
- Correspondence: ; Tel.: +34-653-675-149
| | - Beatriz Monsalve
- Department of Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (B.M.); (B.C.); (M.J.I.); (G.S.); (J.C.I.)
| | - Angela Alonso-Diez
- Department of Animal Medicine, Surgery and Pathology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (A.A.-D.); (P.J.d.A.)
| | - Belén Crespo
- Department of Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (B.M.); (B.C.); (M.J.I.); (G.S.); (J.C.I.)
| | - Maria Jose Illera
- Department of Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (B.M.); (B.C.); (M.J.I.); (G.S.); (J.C.I.)
| | - Paloma Jimena de Andres
- Department of Animal Medicine, Surgery and Pathology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (A.A.-D.); (P.J.d.A.)
| | - Gema Silvan
- Department of Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (B.M.); (B.C.); (M.J.I.); (G.S.); (J.C.I.)
| | - Juan Carlos Illera
- Department of Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (B.M.); (B.C.); (M.J.I.); (G.S.); (J.C.I.)
| |
Collapse
|
5
|
Cruz-Tapias P, Rubiano W, Rondón-Lagos M, Villegas VE, Rangel N. Intrinsic Subtypes and Androgen Receptor Gene Expression in Primary Breast Cancer. A Meta-Analysis. BIOLOGY 2021; 10:biology10090834. [PMID: 34571711 PMCID: PMC8466727 DOI: 10.3390/biology10090834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/29/2021] [Accepted: 08/13/2021] [Indexed: 12/17/2022]
Abstract
The androgen receptor (AR) is frequently expressed in breast cancer (BC), but its association with clinical and biological parameters of BC patients remains unclear. Here, we investigated the association of AR gene expression according to intrinsic BC subtypes by meta-analysis of large-scale microarray transcriptomic datasets. Sixty-two datasets including 10315 BC patients were used in the meta-analyses. Interestingly, AR mRNA level is significantly increased in patients categorized with less aggressive intrinsic molecular subtypes including, Luminal A compared to Basal-like (standardized mean difference, SMD: 2.12; 95% confidence interval, CI: 1.88 to 2.35; p < 0.001) or when comparing Luminal B to Basal-like (SMD: 1.53; CI: 1.33 to 1.72; p < 0.001). The same trend was observed when analyses were performed using immunohistochemistry-based surrogate subtypes. Consistently, the AR mRNA expression was higher in patients with low histological grade (p < 0.001). Furthermore, our data revealed higher levels of AR mRNA in BC patients expressing either estrogen or progesterone receptors (p < 0.001). Together, our findings indicate that high mRNA levels of AR are associated with BC subgroups with the less aggressive clinical features.
Collapse
Affiliation(s)
- Paola Cruz-Tapias
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia; (P.C.-T.); (M.R.-L.)
| | - Wilson Rubiano
- Hospital Universitario Mayor Méderi-Universidad del Rosario, 111411 Bogotá, Colombia;
| | - Milena Rondón-Lagos
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia; (P.C.-T.); (M.R.-L.)
| | - Victoria-E. Villegas
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá 111221, Colombia
- Correspondence: (V.-E.V.); (N.R.); Tel./Fax: +57-1-297-0200 (ext. 4029) (V.-E.V.); +57-1-3185087624 (N.R.)
| | - Nelson Rangel
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
- Correspondence: (V.-E.V.); (N.R.); Tel./Fax: +57-1-297-0200 (ext. 4029) (V.-E.V.); +57-1-3185087624 (N.R.)
| |
Collapse
|
6
|
Carcinogenesis of Triple-Negative Breast Cancer and Sex Steroid Hormones. Cancers (Basel) 2021; 13:cancers13112588. [PMID: 34070471 PMCID: PMC8197527 DOI: 10.3390/cancers13112588] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/19/2021] [Accepted: 05/23/2021] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Triple-negative breast cancer (TNBC) lacks all of three treatment targets (estrogen receptor-α, ER-α; progesterone receptor, PgR; and human epidermal growth factor receptor 2, HER2) and is usually associated with a poor clinical outcome; however, several sex steroid receptors, such as androgen receptor (AR), ER-β, and G-protein-coupled estrogen receptor, are frequently expressed and their biological and clinical importance has been suggested. Despite the structural similarity between sex steroid hormones (androgens and estrogens) or receptors (AR and ER-β), similar signaling mechanisms of these hormones, and the coexistence of these hormones and their receptors in TNBC in a clinical setting, most studies or reviews focused on only one of these receptors, and rarely reviewed them in a comprehensive way. In this review, the carcinogenic or pathobiological role of sex steroid hormones in TNBC is considered, focusing on common and differing features of hormone actions. Abstract Triple-negative breast cancer (TNBC) lacks an effective treatment target and is usually associated with a poor clinical outcome; however, hormone unresponsiveness, which is the most important biological characteristic of TNBC, only means the lack of nuclear estrogenic signaling through the classical estrogen receptor (ER), ER-α. Several sex steroid receptors other than ER-α: androgen receptor (AR), second ER, ER-β, and non-nuclear receptors represented by G-protein-coupled estrogen receptor (GPER), are frequently expressed in TNBC and their biological and clinical importance has been suggested by a large number of studies. Despite the structural similarity between each sex steroid hormone (androgens and estrogens) or each receptor (AR and ER-β), and similarity in the signaling mechanisms of these hormones, most studies or reviews focused on one of these receptors, and rarely reviewed them in a comprehensive way. Considering the coexistence of these hormones and their receptors in TNBC in a clinical setting, a comprehensive viewpoint would be important to correctly understand the association between the carcinogenic mechanism or pathobiology of TNBC and sex steroid hormones. In this review, the carcinogenic or pathobiological role of sex steroid hormones in TNBC is considered, focusing on the common and divergent features of the action of these hormones.
Collapse
|
7
|
Obesity and Androgen Receptor Signaling: Associations and Potential Crosstalk in Breast Cancer Cells. Cancers (Basel) 2021; 13:cancers13092218. [PMID: 34066328 PMCID: PMC8125357 DOI: 10.3390/cancers13092218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/18/2021] [Accepted: 04/03/2021] [Indexed: 12/24/2022] Open
Abstract
Obesity is an increasing health challenge and is recognized as a breast cancer risk factor. Although obesity-related breast cancer mechanisms are not fully understood, this association has been linked to impaired hormone secretion by the dysfunctional obese adipose tissue (hyperplasic and hypertrophic adipocytes). Among these hormones, altered production of androgens and adipokines is observed, and both, are independently associated with breast cancer development. In this review, we describe and comment on the relationships reported between these factors and breast cancer, focusing on the biological associations that have helped to unveil the mechanisms by which signaling from androgens and adipokines modifies the behavior of mammary epithelial cells. Furthermore, we discuss the potential crosstalk between the two most abundant adipokines produced by the adipose tissue (adiponectin and leptin) and the androgen receptor, an emerging marker in breast cancer. The identification and understanding of interactions among adipokines and the androgen receptor in cancer cells are necessary to guide the development of new therapeutic approaches in order to prevent and cure obesity and breast cancer.
Collapse
|
8
|
Laforest S, Pelletier M, Denver N, Poirier B, Nguyen S, Walker BR, Durocher F, Homer NZM, Diorio C, Andrew R, Tchernof A. Estrogens and Glucocorticoids in Mammary Adipose Tissue: Relationships with Body Mass Index and Breast Cancer Features. J Clin Endocrinol Metab 2020; 105:5680713. [PMID: 31853538 PMCID: PMC7065843 DOI: 10.1210/clinem/dgz268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 12/17/2019] [Indexed: 12/16/2022]
Abstract
CONTEXT Adipose tissue is an important site for extragonadal steroid hormone biosynthesis through the expression and activity of P450 aromatase, 11β-hydroxysteroid dehydrogenase (HSD) 1, and 17β-HSDs. The contribution of steroid hormones produced by adjacent adipose tissue for the progression and survival of breast tumors is unknown. OBJECTIVE To quantify estrogens (estradiol, estrone) and glucocorticoids (cortisol, cortisone) in breast adipose tissue from both healthy and diseased women and their relationships with adiposity indices and breast cancer prognostic markers. DESIGN AND SETTING Breast adipose tissue was collected at time of surgery. PATIENTS Pre- and postmenopausal women undergoing partial mastectomy for treatment of breast cancer (n = 17) or reduction mammoplasty (n = 6) were studied. INTERVENTIONS Relative estrogen and glucocorticoid amounts were determined by liquid chromatography tandem mass spectrometry. RESULTS The targeted steroids were reliably detected and quantified in mammary adipose tissues. Women with ER+/PR+ tumor had higher relative estradiol amount than women with ER-/PR- tumor (P < .05). The ratio of estradiol-to-estrone was higher in lean women than in women with a body mass index (BMI) ≥ 25 kg/m2 (P < .05). Mixed-model analyses showed that estradiol, cortisone, and cortisol were negatively associated with tumor size (P < .05). Relationships between glucocorticoids and tumor size remained significant after adjustment for BMI. The cortisol-to-cortisone ratio was negatively associated with tumor stage (P < .05) independently of BMI. CONCLUSIONS We reliably quantified estrogens and glucocorticoids in breast adipose tissue from healthy women and women suffering from breast cancer. Our findings suggest that smaller breast tumors are associated with higher relative amounts of estradiol and cortisol in adipose tissue.
Collapse
Affiliation(s)
- Sofia Laforest
- CHU de Québec-Université Laval Research Center (Endocrinology and Nephrology division), School of Nutrition, Faculty of Agriculture and Food Sciences, Université Laval, Québec, Canada
- Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen’s Medical Research Institute, Edinburgh, UK
| | - Mélissa Pelletier
- CHU de Québec-Université Laval Research Center (Endocrinology and Nephrology division), School of Nutrition, Faculty of Agriculture and Food Sciences, Université Laval, Québec, Canada
- Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
| | - Nina Denver
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen’s Medical Research Institute, Edinburgh, UK
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, University Avenue, Glasgow, UK
| | - Brigitte Poirier
- CHU de Québec-Université Laval Research Center (Oncology division), Université Laval Cancer Research Center and Department of Surgery, Faculty of Medicine, Université Laval, Québec, Canada
- Centre des maladies du sein Deschênes-Fabia, Hôpital Saint-Sacrement, Québec, Canada
| | - Sébastien Nguyen
- CHU de Québec-Université Laval Research Center (Oncology division), Université Laval Cancer Research Center and Department of Surgery, Faculty of Medicine, Université Laval, Québec, Canada
| | - Brian R Walker
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Francine Durocher
- CHU de Québec-Université Laval Research Center (Endocrinology and Nephrology division), Université Laval Cancer Research Center and Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| | - Natalie Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen’s Medical Research Institute, Edinburgh, UK
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Caroline Diorio
- Centre des maladies du sein Deschênes-Fabia, Hôpital Saint-Sacrement, Québec, Canada
- CHU de Québec-Université Laval Research Center (Oncology division), Université Laval Cancer Research Center and Department of Social and Preventive Medicine, Faculty of Medicine, Université Laval, Québec, Canada
| | - Ruth Andrew
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen’s Medical Research Institute, Edinburgh, UK
- University/BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - André Tchernof
- CHU de Québec-Université Laval Research Center (Endocrinology and Nephrology division), School of Nutrition, Faculty of Agriculture and Food Sciences, Université Laval, Québec, Canada
- Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Québec, Canada
- Correspondence and Reprint Requests: André Tchernof, PhD, Institut universitaire de cardiologie et de pneumologie de Québec, 2725 Chemin Ste-Foy, Y4212, Québec, QC, Canada G1V 4G5. E-mail:
| |
Collapse
|
9
|
Inda MA, Blok EJ, Kuppen PJK, Charehbili A, den Biezen-Timmermans EC, van Brussel A, Fruytier SE, Meershoek-Klein Kranenbarg E, Kloet S, van der Burg B, Martens JWM, Sims AH, Turnbull AK, Dixon JM, Verhaegh W, Kroep JR, van de Velde CJH, van de Stolpe A. Estrogen Receptor Pathway Activity Score to Predict Clinical Response or Resistance to Neoadjuvant Endocrine Therapy in Primary Breast Cancer. Mol Cancer Ther 2019; 19:680-689. [PMID: 31727690 DOI: 10.1158/1535-7163.mct-19-0318] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 08/08/2019] [Accepted: 11/08/2019] [Indexed: 11/16/2022]
Abstract
Endocrine therapy is important for management of patients with estrogen receptor (ER)-positive breast cancer; however, positive ER staining does not reliably predict therapy response. We assessed the potential to improve prediction of response to endocrine treatment of a novel test that quantifies functional ER pathway activity from mRNA levels of ER pathway-specific target genes. ER pathway activity was assessed on datasets from three neoadjuvant-treated ER-positive breast cancer patient cohorts: Edinburgh: 3-month letrozole, 55 pre-/2-week/posttreatment matched samples; TEAM IIa: 3- to 6-month exemestane, 49 pre-/28 posttreatment paired samples; and NEWEST: 16-week fulvestrant, 39 pretreatment samples. ER target gene mRNA levels were measured in fresh-frozen tissue (Edinburgh, NEWEST) with Affymetrix microarrays, and in formalin-fixed paraffin-embedded samples (TEAM IIa) with qRT-PCR. Approximately one third of ER-positive patients had a functionally inactive ER pathway activity score (ERPAS), which was associated with a nonresponding status. Quantitative ERPAS decreased significantly upon therapy (P < 0.001 Edinburgh and TEAM IIa). Responders had a higher pretreatment ERPAS and a larger 2-week decrease in activity (P = 0.02 Edinburgh). Progressive disease was associated with low baseline ERPAS (P = 0.03 TEAM IIa; P = 0.02 NEWEST), which did not decrease further during treatment (P = 0.003 TEAM IIa). In contrast, the staining-based ER Allred score was not significantly associated with therapy response (P = 0.2). The ERPAS identified a subgroup of ER-positive patients with a functionally inactive ER pathway associated with primary endocrine resistance. Results confirm the potential of measuring functional ER pathway activity to improve prediction of response and resistance to endocrine therapy.
Collapse
Affiliation(s)
| | - Erik J Blok
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.,Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Ayoub Charehbili
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Sevgi E Fruytier
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Susan Kloet
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Andrew H Sims
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, United Kingdom
| | - Arran K Turnbull
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, United Kingdom.,Edinburgh Breast Unit, Western General Hospital, Edinburgh, United Kingdom
| | - J Michael Dixon
- Edinburgh Breast Unit, Western General Hospital, Edinburgh, United Kingdom
| | | | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | |
Collapse
|
10
|
Creevey L, Bleach R, Madden SF, Toomey S, Bane FT, Varešlija D, Hill AD, Young LS, McIlroy M. Altered Steroid Milieu in AI-Resistant Breast Cancer Facilitates AR Mediated Gene-Expression Associated with Poor Response to Therapy. Mol Cancer Ther 2019; 18:1731-1743. [PMID: 31289138 DOI: 10.1158/1535-7163.mct-18-0791] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/23/2018] [Accepted: 07/03/2019] [Indexed: 11/16/2022]
Abstract
Divergent roles for androgen receptor (AR) in breast cancer have been reported. Following aromatase inhibitor (AI) treatment, the conversion of circulating androgens into estrogens can be diminished by >99%. We wished to establish whether the steroid environment can dictate the role of AR and the implications of this for subsequent therapy. This study utilizes models of AI resistance to explore responsiveness to PI3K/mTOR and anti-AR therapy when cells are exposed to unconverted weak androgens. Transcriptomic alterations driven by androstenedione (4AD) were assessed by RNA-sequencing. AR and estrogen receptor (ER) recruitment to target gene promoters was evaluated using ChIP, and relevance to patient profiles was performed using publicly available data sets. Although BEZ235 showed decreased viability across AI-sensitive and -resistant cell lines, anti-AR treatment elicited a decrease in cell viability only in the AI-resistant model. Serum and glucocorticoid-regulated kinase 3 (SGK3) and cAMP-dependent protein kinase inhibitor β (PKIB) were confirmed to be regulated by 4AD and shown to be mediated by AR; crucially, reexposure to estradiol suppressed expression of these genes. Meta-analysis of transcript levels showed high expression of SGK3 and PKIB to be associated with poor response to endocrine therapy (HR = 2.551, P = 0.003). Furthermore, this study found levels of SGK3 to be sustained in patients who do not respond to AI therapy. This study highlights the importance of the tumor steroid environment. SGK3 and PKIB are associated with poor response to endocrine therapy and could have utility in tailoring therapeutic approaches.
Collapse
Affiliation(s)
- Laura Creevey
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Rachel Bleach
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Stephen F Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Sinead Toomey
- Department of Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Fiona T Bane
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Damir Varešlija
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Arnold D Hill
- Department of Surgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Leonie S Young
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Marie McIlroy
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.
| |
Collapse
|
11
|
Characteristics of adverse events of endocrine therapies among older patients with breast cancer. Support Care Cancer 2019; 27:3813-3822. [DOI: 10.1007/s00520-019-04674-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/26/2018] [Indexed: 11/26/2022]
|
12
|
Bleach R, McIlroy M. The Divergent Function of Androgen Receptor in Breast Cancer; Analysis of Steroid Mediators and Tumor Intracrinology. Front Endocrinol (Lausanne) 2018; 9:594. [PMID: 30416486 PMCID: PMC6213369 DOI: 10.3389/fendo.2018.00594] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/19/2018] [Indexed: 12/16/2022] Open
Abstract
Androgen receptor (AR) is the most widely expressed steroid receptor protein in normal breast tissue and is detectable in approximately 90% of primary breast cancers and 75% of metastatic lesions. However, the role of AR in breast cancer development and progression is mired in controversy with evidence suggesting it can either inhibit or promote breast tumorigenesis. Studies have shown it to antagonize estrogen receptor alpha (ERα) DNA binding, thereby preventing pro-proliferative gene transcription; whilst others have demonstrated AR to take on the mantle of a pseudo ERα particularly in the setting of triple negative breast cancer. Evidence for a potentiating role of AR in the development of endocrine resistant breast cancer has also been mounting with reports associating high AR expression with poor response to endocrine treatment. The resurgence of interest into the function of AR in breast cancer has resulted in various emergent clinical trials evaluating anti-AR therapy and selective androgen receptor modulators in the treatment of advanced breast cancer. Trials have reported varied response rates dependent upon subtype with overall clinical benefit rates of ~19-29% for anti-androgen monotherapy, suggesting that with enhanced patient stratification AR could prove efficacious as a breast cancer therapy. Androgens and AR have been reported to facilitate tumor stemness in some cancers; a process which may be mediated through genomic or non-genomic actions of the AR, with the latter mechanism being relatively unexplored in breast cancer. Steroidogenic ligands of the AR are produced in females by the gonads and as sex-steroid precursors secreted from the adrenal glands. These androgens provide an abundant reservoir from which all estrogens are subsequently synthesized and their levels are undiminished in the event of standard hormonal therapeutic intervention in breast cancer. Steroid levels are known to be altered by lifestyle factors such as diet and exercise; understanding their potential role in dictating the function of AR in breast cancer development could therefore have wide-ranging effects in prevention and treatment of this disease. This review will outline the endogenous biochemical drivers of both genomic and non-genomic AR activation and how these may be modulated by current hormonal therapies.
Collapse
Affiliation(s)
| | - Marie McIlroy
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
13
|
Africander D, Storbeck KH. Steroid metabolism in breast cancer: Where are we and what are we missing? Mol Cell Endocrinol 2018; 466:86-97. [PMID: 28527781 DOI: 10.1016/j.mce.2017.05.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 05/08/2017] [Accepted: 05/16/2017] [Indexed: 12/12/2022]
Abstract
It is well-known that breast cancer is hormone-dependent and that steroid hormones exert their mitogenic effects by binding to estrogen, progesterone and androgen receptors. Vital to our understanding and treatment of this malignancy, is the local metabolism of steroid hormones in breast cancer tissue. This review summarises our current knowledge on steroid producing pathways in the adrenal, ovary and breast, while focussing on the availability of specific circulating hormone precursors and steroidogenic enzymes involved in the local synthesis and metabolism of steroid hormones in the breast. Consequently, we highlight alternate pathways that may be instrumental in the etiology of breast cancer.
Collapse
Affiliation(s)
- Donita Africander
- Department of Biochemistry, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch 7600, South Africa.
| |
Collapse
|
14
|
Moon JY, McNamara KM, Lee JJ, Chung BC, Sasano H, Choi MH. Improved detectability of sex steroids from frozen sections of breast cancer tissue using GC-triple quadrupole-MS. J Steroid Biochem Mol Biol 2018; 178:185-192. [PMID: 29269263 DOI: 10.1016/j.jsbmb.2017.12.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/10/2017] [Accepted: 12/14/2017] [Indexed: 11/18/2022]
Abstract
Sex steroids in clinical endocrinology have been mainly investigated with peripheral blood and urine samples, while there is limited information regarding the local levels within tissues. To improve analytical properties of sex steroids from trace amounts of tissue samples, two-phase extractive ethoxycarbonlyation and subsequent pentafluoropropionyl derivatization coupled to gas chromatography-tandem mass spectrometry (GC-MS/MS) was developed. The optimized analytical conditions led to excellent chromatographic separation of 15 estrogens, 6 androgens, and 2 progestins. The quantitative results were calculated based on in-house control samples as the steroid-free tissues, and the precision and accuracy were 4.2%-26.8% and 90.8%-116.4%, respectively. The on-column limit of quantification was from 180 fg to 0.5 pg for androgens and estrogens, and 1.25 pg for progestins, which were found to be linear (r2 > 0.990). The validated method was then applied to quantify 7 sex steroids from three 100-μm-thick frozen breast tissue slices from postmenopausal patients with breast cancer. This is the first report on the improved GC-MS/MS method for the detection of androgens and pregnenolone from breast cancer tissues, and it can be a useful technique to measure the local levels of sex steroids, thus, enhancing our understanding of the pathophysiological significances of steroidogenesis.
Collapse
Affiliation(s)
- Ju-Yeon Moon
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Keely May McNamara
- Department of Pathology, Tohoku University School of Medicine, Sendai, 980-8575, Japan
| | - Jung-Jin Lee
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Bong Chul Chung
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hironobu Sasano
- Department of Pathology, Tohoku University School of Medicine, Sendai, 980-8575, Japan
| | - Man Ho Choi
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| |
Collapse
|
15
|
Plaza-Parrochia F, Romero C, Valladares L, Vega M. Endometrium and steroids, a pathologic overview. Steroids 2017; 126:85-91. [PMID: 28827068 DOI: 10.1016/j.steroids.2017.08.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/07/2017] [Accepted: 08/12/2017] [Indexed: 11/27/2022]
Abstract
Normal endometrial function requires of cell proliferation and differentiation; therefore, disturbances in these processes could lead to pathological entities such as hyperplasia and endometrial adenocarcinoma, where cell proliferation is increased. The development of these pathologies is highly related to alterations in the levels and/or action of sexual steroids. In the present review, it has been analyzed how steroids, particularly estrogens, androgens and progestagens are involved in the etiopathogenesis of hyperplasia and endometrial endometrioid adenocarcinoma. The emphasis is given on pathological and pharmacological conditions that are presented as risk factors for endometrial pathologies, such as obesity, polycystic ovarian syndrome and hormone replacement postmenopausal women therapy, among others. Steroids alterations may promote changes at molecular level that enhance the development of hyperplasia and endometrioid cancer. In fact, there are solid data that indicate that estrogens stimulate cell-proliferation in this tissue; meanwhile, progestagens are able to stop cell proliferation and to increase differentiation. Nevertheless, the role of androgens is less clear, since there is contradictory information. It is most likely that the major contribution of steroids to the development of cell proliferation pathologies in endometria would be in early stages, where there is a high sensitivity to these molecules. This phenomenon is present even in stages previous to the occurrence of hyperplasia, like in the condition of polycystic ovarian syndrome, where the endometria have a greater sensitivity to steroids and high expression of cell cycle molecules. These abnormalities would contribute to the pathogenesis of hyperplasia and then in the progression to endometrioid adenocarcinoma.
Collapse
Affiliation(s)
- Francisca Plaza-Parrochia
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont # 999, Santiago, Chile.
| | - Carmen Romero
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont # 999, Santiago, Chile
| | - Luis Valladares
- Institute of Nutrition and Food Technology, University of Chile, Macul #5540, Chile
| | - Margarita Vega
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont # 999, Santiago, Chile.
| |
Collapse
|
16
|
Caceres S, Monsalve B, Peña L, de Andres PJ, Alonso-Diez A, Illera MJ, Woodward WA, Reuben JM, Silvan G, Illera JC. In vitro and in vivo effect of flutamide on steroid hormone secretion in canine and human inflammatory breast cancer cell lines. Vet Comp Oncol 2017; 16:148-158. [PMID: 28589573 DOI: 10.1111/vco.12324] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/12/2017] [Accepted: 05/02/2017] [Indexed: 12/22/2022]
Abstract
The aim was to study the effects of flutamide on cell proliferation, in vivo tumour growth and steroid production in canine and human IBC cell lines. IPC-366 and SUM149 cell cultures were exposed to flutamide concentrations for 72 hours. Additionally, IPC-366 and SUM149 xenotransplanted mice were treated subcutaneously with flutamide 3 times a week for 2 weeks. Steroid hormones determination in culture media, serum and tumour homogenates (pregnenolone, progesterone, androstenedione, testosterone, dihydrotestosterone, 17β-oestradiol and oestrone sulphate) were assayed by EIA. in vitro cell proliferation percentages showed a decrease in all flutamide dosages in IPC-366 and SUM149. in vivo flutamide reduced tumour size by 55% to 65%, and metastasis rates decreased. In treated groups, androgen levels in culture media, serum and tumour homogenates were increased as oestrogen levels decreased. These results suggest that flutamide treatment inhibits cell proliferation and promotes tumour reduction by increasing androgen levels and also support future therapy approaches.
Collapse
Affiliation(s)
- S Caceres
- Department of Animal Physiology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Madrid, Spain
| | - B Monsalve
- Department of Animal Physiology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Madrid, Spain
| | - L Peña
- Department of Animal Medicine Surgery and Pathology, School of Veterinary Medicine. Complutense University of Madrid (UCM), Madrid, Spain
| | - P J de Andres
- Department of Animal Medicine Surgery and Pathology, School of Veterinary Medicine. Complutense University of Madrid (UCM), Madrid, Spain
| | - A Alonso-Diez
- Department of Animal Medicine Surgery and Pathology, School of Veterinary Medicine. Complutense University of Madrid (UCM), Madrid, Spain
| | - M J Illera
- Department of Animal Physiology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Madrid, Spain
| | - W A Woodward
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - J M Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - G Silvan
- Department of Animal Physiology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Madrid, Spain
| | - J C Illera
- Department of Animal Physiology, School of Veterinary Medicine, Complutense University of Madrid (UCM), Madrid, Spain
| |
Collapse
|
17
|
Dudenkov TM, Ingle JN, Buzdar AU, Robson ME, Kubo M, Ibrahim-Zada I, Batzler A, Jenkins GD, Pietrzak TL, Carlson EE, Barman P, Goetz MP, Northfelt DW, Moreno-Aspita A, Williard CV, Kalari KR, Nakamura Y, Wang L, Weinshilboum RM. SLCO1B1 polymorphisms and plasma estrone conjugates in postmenopausal women with ER+ breast cancer: genome-wide association studies of the estrone pathway. Breast Cancer Res Treat 2017; 164:189-199. [PMID: 28429243 DOI: 10.1007/s10549-017-4243-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/10/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Estrone (E1), the major circulating estrogen in postmenopausal women, promotes estrogen-receptor positive (ER+) breast tumor growth and proliferation. Two major reactions contribute to E1 plasma concentrations, aromatase (CYP19A1) catalyzed E1 synthesis from androstenedione and steroid sulfatase (STS) catalyzed hydrolysis of estrone conjugates (E1Cs). E1Cs have been associated with breast cancer risk and may contribute to tumor progression since STS is expressed in breast cancer where its activity exceeds that of aromatase. METHODS We performed genome-wide association studies (GWAS) to identify SNPs associated with variation in plasma concentrations of E1Cs, E1, and androstenedione in 774 postmenopausal women with resected early-stage ER+ breast cancer. Hormone concentrations were measured prior to aromatase inhibitor therapy. RESULTS Multiple SNPs in SLCO1B1, a gene encoding a hepatic influx transporter, displayed genome-wide significant associations with E1C plasma concentrations and with the E1C/E1 ratio. The top SNP for E1C concentrations, rs4149056 (p = 3.74E-11), was a missense variant that results in reduced transporter activity. Patients homozygous for the variant allele had significantly higher average E1C plasma concentrations than did other patients. Furthermore, three other SLCO1B1 SNPs, not in LD with rs4149056, were associated with both E1C concentrations and the E1C/E1 ratio and were cis-eQTLs for SLCO1B3. GWAS signals of suggestive significance were also observed for E1, androstenedione, and the E1/androstenedione ratio. CONCLUSION These results suggest a mechanism for genetic variation in E1C plasma concentrations as well as possible SNP biomarkers to identify ER+ breast cancer patients for whom STS inhibitors might be of clinical value.
Collapse
Affiliation(s)
- Tanda M Dudenkov
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - James N Ingle
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Aman U Buzdar
- Department of Breast Oncology, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Mark E Robson
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
| | - Irada Ibrahim-Zada
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.,University of Colorado, Denver, USA
| | - Anthony Batzler
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Gregory D Jenkins
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - Erin E Carlson
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Poulami Barman
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Matthew P Goetz
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Krishna R Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Yusuke Nakamura
- Department of Medicine, School of Medicine, University of Chicago, Chicago, IL, USA
| | - Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Richard M Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
18
|
Honma N, Saji S, Mikami T, Yoshimura N, Mori S, Saito Y, Murayama S, Harada N. Estrogen-Related Factors in the Frontal Lobe of Alzheimer's Disease Patients and Importance of Body Mass Index. Sci Rep 2017; 7:726. [PMID: 28389656 PMCID: PMC5429714 DOI: 10.1038/s41598-017-00815-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 03/16/2017] [Indexed: 01/22/2023] Open
Abstract
Estrogens play a physiologically important role in the brain, but controversies exist regarding the association between Alzheimer’s disease (AD) and estrogens. Estrogen-related factors were comprehensively examined in frontal lobe tissues from autopsied AD patients, and compared with controls. Concentrations of estrogens, expression of estrogen receptors (ERs), and estrogen-metabolizing enzymes (EMEs) which are important for determining the peripheral estrogen concentrations, were examined using liquid chromatography tandem mass spectrometry, immunohistochemistry, and quantitative real-time PCR, respectively. Body mass index (BMI), known to correlate with the serum estrogen concentrations, was also taken into consideration. There were no significant differences in estrogen concentrations or each EME level between the two groups in both the cortex and white matter, whereas glial nuclear ER-β expression was significantly lower in white matter from the AD group than the control group (Allred score, 3.2 ± 0.3 and 6.5 ± 0.3, respectively. P < 0.0001). Estrogen concentrations were found to closely correlate with BMI, particularly in controls. ER-β loss in the white matter from the AD group suggests the necessity of studying the effects of estrogens on glias as well as neurons in the etiology of AD. The correlation between BMI and estrogen concentrations in the frontal lobe suggests the importance of non-brain sources of estrogens.
Collapse
Affiliation(s)
- Naoko Honma
- Department of Pathology, Toho University School of Medicine, Omori-Nishi 5-21-16, Ota-ku, Tokyo, 143-8540, Japan.
| | - Shigehira Saji
- Department of Medical Oncology, Fukushima Medical University, Hikariga-oka 1, Fukushima City, Fukushima, 960-1295, Japan
| | - Tetuo Mikami
- Department of Pathology, Toho University School of Medicine, Omori-Nishi 5-21-16, Ota-ku, Tokyo, 143-8540, Japan
| | - Noriko Yoshimura
- Department of Biochemistry, Fujita Health University School of Medicine, Dengakugakubo 1-98, Kutsukake-cho, Toyoake, 470-1192, Japan
| | - Seijiro Mori
- Department of Internal Medicine, Tokyo Metropolitan Geriatric Hospital, Sakaecho 35-2, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Yuko Saito
- Department of Pathology and Laboratory Medicine, National Center Hospital, National Center of Neurology and Psychiatry, Ogawa-Higashi 4-1-1, Kodaira, Tokyo, 187-8551, Japan
| | - Shigeo Murayama
- Department of Neuropathology, Tokyo Metropolitan Institute of Gerontology, Sakaecho 35-2, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Nobuhiro Harada
- Department of Biochemistry, Fujita Health University School of Medicine, Dengakugakubo 1-98, Kutsukake-cho, Toyoake, 470-1192, Japan
| |
Collapse
|
19
|
Overcoming aromatase inhibitor resistance in breast cancer: possible mechanisms and clinical applications. Breast Cancer 2017; 25:379-391. [PMID: 28389808 DOI: 10.1007/s12282-017-0772-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/28/2017] [Indexed: 10/19/2022]
Abstract
Estrogen plays crucial roles in the progression of hormone-dependent breast cancers through activation of nuclear estrogen receptor α (ER). Estrogen is produced locally from circulating inactive steroids and adrenal androgens in postmenopausal women. However, conversion by aromatase is a rate-limiting step in intratumoral estrogen production in breast cancer. Aromatase inhibitors (AIs) inhibit the growth of hormone-dependent breast cancers by blocking the conversion of adrenal androgens to estrogen and by unmasking the inhibitory effect of androgens, acting via the androgen receptor (AR). AIs are thus a standard treatment option for postmenopausal hormone-dependent breast cancer. However, although initial use of AIs provides substantial clinical benefit, some breast cancer patients relapse because of the acquisition of AI resistance. A better understanding of the mechanisms of AI resistance may contribute to the development of new therapeutic strategies and aid in the search for new therapeutic targets and agents. We have investigated AI-resistance mechanisms and established six AI-resistant cell lines. Some of them exhibit estrogen depletion-resistance properties via constitutive ER-activation or ER-independent growth signaling. We examined how breast cancer cells can adapt to estrogen depletion and androgen superabundance. Estrogen and estrogenic androgen produced independently from aromatase contributed to cell proliferation in some of these cell lines, while another showed AR-dependent cell proliferation. Based on these findings, currently proposed AI-resistance mechanisms include an aromatase-independent estrogen-producing pathway, estrogen-independent ER function, and ER-independent growth signaling. This review summarizes several hypotheses of AI-resistance mechanisms and discusses how existing or novel therapeutic agents may be applied to treat AI-resistant breast cancers.
Collapse
|
20
|
Associations of obesity and physical activity with serum and intratumoral sex steroid hormone levels among postmenopausal women with breast cancer: analysis of paired serum and tumor tissue samples. Breast Cancer Res Treat 2017; 162:115-125. [PMID: 28044214 DOI: 10.1007/s10549-016-4094-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/23/2016] [Indexed: 12/14/2022]
Abstract
PURPOSE It has been hypothesized that intratumoral estrogens may play important roles in the growth of breast cancer. However, few studies have investigated such intratumoral hormones, or their association with risk factors of breast cancer. METHODS In this cross-sectional study, hormone levels in paired serum and tumor tissue samples from 146 postmenopausal women with breast cancer were measured by liquid chromatography-tandem mass spectrometry and compared between estrogen/progesterone (ER/PgR) subtypes. The associations of risk factors including body mass index (BMI) and other lifestyle factors with these hormone levels were investigated using analysis of covariance. RESULTS The level of estradiol (E2) in tumor tissue was extremely high in women with ER+ (geometric mean 95.6 pg/g) relative to women with ER-/PgR- (8.9 pg/g), whereas serum E2 level did not differ much between the two groups (3.1 and 2.8 pg/ml, respectively). Serum levels of precursors for E2, including testosterone (T) and androstenedione (Adione), and tissue Adione level, were high among women with ER+. After adjustment for confounding variables, BMI was found to be positively associated with tissue levels of E2, estrone (E1), T, and Adione among women with ER+ (P trend < 0.0001 for E2; 0.0016 for E1; 0.0002 for T; and 0.03 for Adione). CONCLUSION The data suggest that tissue E2 is related to the growth of receptor-positive breast cancer and that risk factors such as BMI affect tissue levels of E2 and its precursors. Understanding of hormonal environments within tumor tissue may be important for elucidating hormonal etiology of breast cancer and improving the prognosis of patients.
Collapse
|
21
|
Plaza-Parrochia F, Oróstica L, García P, Vera C, Romero C, Valladares L, Vega M. Molecular Mechanisms of Androstenediol in the Regulation of the Proliferative Process of Human Endometrial Cells. Reprod Sci 2016; 24:1079-1087. [DOI: 10.1177/1933719116678689] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Francisca Plaza-Parrochia
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile Clinical Hospital, Santiago, Chile
| | - Lorena Oróstica
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile Clinical Hospital, Santiago, Chile
| | - Paula García
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile Clinical Hospital, Santiago, Chile
| | - Carolina Vera
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile Clinical Hospital, Santiago, Chile
| | - Carmen Romero
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile Clinical Hospital, Santiago, Chile
| | - Luis Valladares
- Institute of Nutrition and Food Technology, University of Chile, Macul, Chile
| | - Margarita Vega
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile Clinical Hospital, Santiago, Chile
| |
Collapse
|
22
|
McNamara KM, Oguro S, Omata F, Kikuchi K, Guestini F, Suzuki K, Yang Y, Abe E, Hirakawa H, Brown KA, Takanori I, Ohuchi N, Sasano H. The presence and impact of estrogen metabolism on the biology of triple-negative breast cancer. Breast Cancer Res Treat 2016; 161:213-227. [DOI: 10.1007/s10549-016-4050-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/05/2016] [Indexed: 11/25/2022]
|
23
|
Diep CH, Ahrendt H, Lange CA. Progesterone induces progesterone receptor gene (PGR) expression via rapid activation of protein kinase pathways required for cooperative estrogen receptor alpha (ER) and progesterone receptor (PR) genomic action at ER/PR target genes. Steroids 2016; 114:48-58. [PMID: 27641443 PMCID: PMC5068826 DOI: 10.1016/j.steroids.2016.09.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 08/30/2016] [Accepted: 09/02/2016] [Indexed: 12/12/2022]
Abstract
Progesterone Receptors (PRs) are critical effectors of estrogen receptor (ER) signaling required for mammary gland development and reproductive proficiency. In breast and reproductive tract malignancies, PR expression is a clinical prognostic marker of ER action. While estrogens primarily regulate PR expression, other factors likely contribute to a dynamic range of receptor expression across diverse tissues. In this study, we identified estrogen-independent but progestin (R5020)-dependent regulation of ER target genes including PGR in ER+/PR+ cancer cell lines. R5020 (10nM-10μM range) induced dose-dependent PR mRNA and protein expression in the absence of estrogen but required both PR and ERα. Antagonists of either PR (RU486, onapristone) or ERα (ICI 182,780) attenuated R5020 induction of TFF1, CTSD, and PGR. Chromatin immunoprecipitation (ChIP) assays performed on ER+/PR+ cells demonstrated that both ERα and PR were recruited to the same ERE/Sp1 site-containing region of the PGR proximal promoter in response to high dose progestin (10μM). Recruitment of ERα and PR to chromatin and subsequent PR mRNA induction were dependent upon rapid activation of MAPK/ERK and AKT; inhibition of these kinase pathways via U0126 or LY294002 blocked these events. Overall, we have identified a novel mechanism of ERα activation initiated by rapid PR-dependent kinase pathway activation and associated with phosphorylation of ERα Ser118 for estrogen-independent but progestin-dependent ER/PR cross talk. These studies may provide insight into mechanisms of persistent ER-target gene expression during periods of hormone (i.e. estrogen) ablation and suggest caution following prolonged treatment with aromatase or CYP17 inhibitors (i.e. contexts when progesterone levels may be abnormally elevated).
Collapse
Affiliation(s)
- Caroline H Diep
- Department of Medicine (Division of Hematology, Oncology, and Transplantation), University of Minnesota, Minneapolis, MN 55455, United States.
| | - Hannah Ahrendt
- Department of Medicine (Division of Hematology, Oncology, and Transplantation), University of Minnesota, Minneapolis, MN 55455, United States.
| | - Carol A Lange
- Department of Medicine (Division of Hematology, Oncology, and Transplantation), University of Minnesota, Minneapolis, MN 55455, United States; Department of Pharmacology, and Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, United States.
| |
Collapse
|
24
|
Plaza-Parrochia F, Poblete C, Gabler F, Carvajal R, Romero C, Valladares L, Vega M. Expression of steroid sulfated transporters and 3β-HSD activity in endometrium of women having polycystic ovary syndrome. Steroids 2015; 104:189-95. [PMID: 26450365 DOI: 10.1016/j.steroids.2015.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/02/2015] [Accepted: 10/03/2015] [Indexed: 12/15/2022]
Abstract
Intracrinology mechanism involves the metabolism of steroids in peripheral tissues, such as DHEA, to molecules with estrogenic or androgenic activity. Proliferation rate of endometria from Polycystic Ovary Syndrome women (PCOS) is increased, favoring hyperplasia development. Besides, in endometria from PCOS-women the synthesis of androst-5-ene-3β,17β-diol (androstenediol), an estrogenic molecule, is enhanced concomitantly to increased cellular proliferation. DHEA, the major intracrinological precursor, circulates mainly in its sulfated form and requires transporters for cell intake, that belong to the families of organic anion transporting polypeptides (OATP) and organic anion transporters (OAT). The aim of this study was to determine protein levels and activity of sulfated steroid transporters OATP2B1, OATP3A1, OATP4A1 and OAT4 in endometria from control and PCOS-women and to evaluate the activity of the enzyme 3β-HSD. Levels of transporters were done by RT-PCR (OAT4 only) and Western-blot (WB). Additionally, in primary culture cells stimulated with steroids, protein levels by WB and uptake of tritiated DHEAS, were evaluated; 3β-HSD activity was assessed using radiolabel substrate. PCOS-endometrium had higher levels of OATP2B1 and OATP4A1 than CE (p<0.05); decreased OATP4A1 levels were found in androstenediol or testosterone-stimulated cells. Accordingly, the entry of DHEAS to cells was lower in cells stimulated with testosterone (p<0.05); 3β-HSD-activity was similar in control and PCOS-endometria. Therefore, this study describes that steroids can modulate the expression and activity of transporters of OATPs-family in human endometria and that some transporter levels are increased in PCOS-endometria, suggesting a potential role in the pathogenesis of endometrial hyperplasia of these patients.
Collapse
Affiliation(s)
- Francisca Plaza-Parrochia
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont #999, Santiago, Chile
| | - Cristian Poblete
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont #999, Santiago, Chile
| | - Fernando Gabler
- Department of Pathology, School of Medicine, University of Chile, San Borja Arriarán Clinical Hospital, Santa Rosa #1234, Chile
| | - Rodrigo Carvajal
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont #999, Santiago, Chile
| | - Carmen Romero
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont #999, Santiago, Chile
| | - Luis Valladares
- Institute of Nutrition and Food Technology, University of Chile, Macul #5540, Chile
| | - Margarita Vega
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont #999, Santiago, Chile.
| |
Collapse
|
25
|
Honma N, Hosoi T, Arai T, Takubo K. Estrogen and cancers of the colorectum, breast, and lung in postmenopausal women. Pathol Int 2015; 65:451-9. [PMID: 26126901 DOI: 10.1111/pin.12326] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 05/28/2015] [Indexed: 01/01/2023]
Abstract
As estrogens play an important role in maintaining physiological function in various organs, the estrogen decrease after menopause is thought to cause various diseases frequently observed in postmenopausal or elderly women. With the aging of society and a decrease in infectious or vascular diseases, neoplasms have now become the most frequent cause of death in Japan. Cancers of the colorectum, breast, and lung have been rapidly increasing both in incidence and death, especially among postmenopausal women. Interestingly, all three of these cancers are associated with estrogens. In premenopausal women, ovarian estrogens plays major roles in the female reproductive organs through the classic estrogen receptor, ER-α. In postmenopausal women, however, estrogens produced/activated by peripherally localized estrogen-metabolizing enzymes such as aromatase, which converts androgen into estrogens, are thought to play physiologically and pathobiologically important roles in various organs through second ER, namely ER-β, distributing systemically. In this article, the association of estrogens with these cancers in postmenopausal or elderly women are reviewed, especially focusing on the role of ER-β and peripheral estrogen metabolism. The possibility of prevention or treatment of these diseases through estrogenic control is also discussed.
Collapse
Affiliation(s)
- Naoko Honma
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Takayuki Hosoi
- Kenkoin Clinic, Institute for Preventive Medicine, Kenkoin Medical Corporation, Tokyo, Japan
| | - Tomio Arai
- Department of Pathology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Kaiyo Takubo
- Research Team for Geriatric Pathology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
26
|
Stanczyk FZ, Mathews BW, Sherman ME. Relationships of sex steroid hormone levels in benign and cancerous breast tissue and blood: A critical appraisal of current science. Steroids 2015; 99:91-102. [PMID: 25554581 DOI: 10.1016/j.steroids.2014.12.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 12/10/2014] [Indexed: 10/24/2022]
Abstract
A systematic review of the literature on sex steroid measurement in breast tissue identified only 19 articles meeting the following criteria: menopausal status given; steroids measured in tissue homogenates by conventional RIA with a purification step or by mass spectrometry; and values reported per g tissue or per g protein. Twelve articles were analyzed in detail for: ratios of sex steroid hormone levels in cancerous or benign tissues to blood levels, stratified by menopausal status; ratios between the different hormone levels within tissues or within blood; and difference in these ratios between tissue and blood compartments. Estrogen and androgen concentrations varied greatly in benign and cancerous tissues and in blood between individuals. Postmenopausal, but not premenopausal, estradiol concentrations were significantly higher in cancerous compared to benign breast tissue. The estradiol/estrone ratio was lowest in premenopausal benign tissue, and substantially higher in premenopausal cancerous tissue and postmenopausal benign and cancerous tissues. Estradiol and estrone levels were considerably higher in tissue than in plasma in both premenopausal and postmenopausal women. Androgen levels were generally higher in the benign than the cancerous tissue, and tissue androgen levels were higher than in plasma, suggesting in situ aromatization of androgens to estrogens in breast cancer tissue. Limited available data on levels of hydroxylated estrogens in breast tissue compared to corresponding levels in plasma or urine were reviewed, but due to the paucity of studies no conclusions can presently be drawn regarding the relationship of the 2-hydroxyestrone:16α-hydroxyestrone ratio to breast cancer risk and genotoxic effects of 4-hydroxylated estrogens. Finally, data on hormone levels in breast adipose tissue were analyzed; high levels of androstenedione and testosterone and significant estrone and estradiol levels in breast adipocytes from postmenopausal breast cancer patients are consistent with an obesity-inflammation-aromatase axis occurring locally in breast tissue. The controversies regarding the source of intratumoral estrogens in the breast are summarized.
Collapse
Affiliation(s)
- Frank Z Stanczyk
- Departments of Obstetrics and Gynecology, and Preventive Medicine, University of Southern California Keck School of Medicine, 1321 N. Mission Rd., Los Angeles, CA 90033, USA.
| | - Brett W Mathews
- University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Mark E Sherman
- National Cancer Institute, Division of Cancer Prevention, 9609 Medical Center Drive, Bethesda, MD 20892, USA
| |
Collapse
|
27
|
Heublein S, Mayr D, Meindl A, Angele M, Gallwas J, Jeschke U, Ditsch N. Thyroid Hormone Receptors Predict Prognosis in BRCA1 Associated Breast Cancer in Opposing Ways. PLoS One 2015; 10:e0127072. [PMID: 26029931 PMCID: PMC4451081 DOI: 10.1371/journal.pone.0127072] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 04/11/2015] [Indexed: 12/02/2022] Open
Abstract
Since BRCA1 associated breast cancers are frequently classified as hormone receptor negative or even triple negative, the application of endocrine therapies is rather limited in these patients. Like hormone receptors that bind to estrogen or progesterone, thyroid hormone receptors (TRs) are members of the nuclear hormone receptor superfamily. TRs might be interesting biomarkers - especially in the absence of classical hormone receptors. The current study aimed to investigate whether TRs may be specifically expressed in BRCA1 associated cancer cases and whether they are of prognostic significance in these patients as compared to sporadic breast cancer cases. This study analyzed TRα and TRβ immunopositivity in BRCA1 associated (n = 38) and sporadic breast cancer (n = 86). Further, TRs were studied in MCF7 (BRCA1 wildtype) and HCC3153 (BRCA1 mutated) cells. TRβ positivity rate was significantly higher in BRCA1 associated as compared to sporadic breast cancers (p = 0.001). The latter observation remained to be significant when cases that had been matched for clinicopathological criteria were compared (p = 0.037). Regarding BRCA1 associated breast cancer cases TRβ positivity turned out to be a positive prognostic factor for five-year (p = 0.007) and overall survival (p = 0.026) while TRα positivity predicted reduced five-year survival (p = 0.030). Activation of TRβ resulted in down-modulation of CTNNB1 while TRα inhibition reduced cell viability in HCC3153. However, only BRCA1 wildtype MCF7 cells were capable of rapidly degrading TRα1 in response to T3 stimulation. Significantly, this study identified TRβ to be up-regulated in BRCA1 associated breast cancer and revealed TRs to be associated with patients’ prognosis. TRs were also found to be expressed in triple negative BRCA1 associated breast cancer. Further studies need to be done in order to evaluate whether TRs may become interesting targets of endocrine therapeutic approaches, especially when tumors are triple-negative.
Collapse
Affiliation(s)
- Sabine Heublein
- Department of Obstetrics and Gynecology, Ludwig-Maximilians-University of Munich, Munich, Germany
- * E-mail:
| | - Doris Mayr
- Department of Pathology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Alfons Meindl
- Department of Obstetrics and Gynecology, Technical University of Munich, Munich, Germany
| | - Martin Angele
- Department of Surgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Julia Gallwas
- Department of Obstetrics and Gynecology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Nina Ditsch
- Department of Obstetrics and Gynecology, Ludwig-Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
28
|
McNamara KM, Sasano H. The intracrinology of breast cancer. J Steroid Biochem Mol Biol 2015; 145:172-8. [PMID: 24751707 DOI: 10.1016/j.jsbmb.2014.04.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/08/2014] [Accepted: 04/09/2014] [Indexed: 02/01/2023]
Abstract
The importance of intracrinology, or in situ production of steroids from circulating precursors, in breast cancer has been firmly established in estrogen actions on postmenopausal patients. Expression levels of various steroid synthesizing and/or metabolizing enzymes have been examined in human breast cancer tissues by a number of groups. The enzymes examined include those capable of converting circulating DHEA-S to sex steroids (STS and 3βHSDΔ4-5 isomerase), the group of enzymes that modulate the strength of both androgens and estrogens (17βHSD family) as well as the androgenic 5αR enzymes and the estrogenic aromatase enzyme. In addition to these DHEA-related metabolism pathways, other intracrine pathways involving progesterone and cholesterol have also been examined. Some risk factors of breast cancer development, including obesity, have also been postulated to interact with steroid metabolising pathways. In this review, we aimed to summarise the current state of knowledge regarding intracrine metabolism including expression levels of various enzymes and receptors, focusing particularly upon the importance of the production of biologically potent steroids from circulating sulfated precursors such as DHEA-S. In addition, we attempted to summarise the factors, both steroidal and non-steroidal, involved in the regulation of these enzymes and propose future directions for research in this particular field. The concept of intracrinology was first proposed over 20 years ago but there still remain many unanswered questions which could open new horizons for the understanding of intracrine metabolism in the breast. This article is part of a Special Issue entitled 'Essential role of DHEA'.
Collapse
Affiliation(s)
- Keely May McNamara
- Department of Pathology, Tohoku University School of Medicine, Sendai, Miyagi, Japan.
| | - Hironobu Sasano
- Department of Pathology, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
29
|
Plaza-Parrochia F, Bacallao K, Poblete C, Gabler F, Carvajal R, Romero C, Valladares L, Vega M. The role of androst-5-ene-3β,17β-diol (androstenediol) in cell proliferation in endometrium of women with polycystic ovary syndrome. Steroids 2014; 89:11-9. [PMID: 25065586 DOI: 10.1016/j.steroids.2014.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/04/2014] [Accepted: 07/09/2014] [Indexed: 02/07/2023]
Abstract
Women with polycystic ovary syndrome (PCOS) show high prevalence of endometrial hyperplasia and adenocarcinoma. Endometrial proliferation is increased, evaluated by high levels of Ki67 (cell cycle marker) and low levels of p27 (negative regulator of cell cycle). Nevertheless, endometrial changes in cyclin D1 (positive regulator of cell cycle) in PCOS-women are not described. Androst-5-ene-3β,17β-diol (androstenediol), steroid with estrogenic activity present in endometria, could be related to increased endometrial cell proliferation. The objective of this study was to determine protein content of cyclin D1 and androstenediol levels in endometria from PCOS and control-women and to evaluate the possible mechanism favoring cell proliferation associated with hormonal characteristics of patients. Therefore, cyclin D1 protein content in PCOS-women and control-endometrial tissue were assessed by western blot and immunohistochemistry. The androstenediol levels were evaluated by ELISA. To further analyze the effect of steroids (androstenediol, 17β-estradiol, testosterone) in cell proliferation, levels of proteins cyclin D1, p27 and Ki67 were evaluated in an in vitro model of stromal endometrial cells T-HESC and St-T1b. An increase in cyclin D1 and androstenediol was observed in tissues from PCOS-women relative to control group (p<0.05). In the in vitro model, androstenediol exerted increase in cyclin D1 (p<0.05) and a decrease in p27 protein level (p<0.05), while Ki67 in St-T1b cells increased under this stimulus (p<0.05). Testosterone produces opposite effects in the levels of the above markers (p<0.05). Therefore, the hormonal imbalance associated with this syndrome could alter endometrial tissue homeostasis, promoting cell proliferation. Androstenediol is a molecule that could be involved by stimulating proliferation, whereas testosterone elicits a role of cell cycle repressor.
Collapse
Affiliation(s)
- Francisca Plaza-Parrochia
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont # 999, Santiago, Chile
| | - Ketty Bacallao
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont # 999, Santiago, Chile
| | - Cristian Poblete
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont # 999, Santiago, Chile
| | - Fernando Gabler
- Department of Pathology, School of Medicine, University of Chile, San Borja Arriarán Clinical Hospital, Santa Rosa #1234, Chile
| | - Rodrigo Carvajal
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont # 999, Santiago, Chile
| | - Carmen Romero
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont # 999, Santiago, Chile
| | - Luis Valladares
- Institute of Nutrition and Food Technology, University of Chile, Macul #5540, Chile
| | - Margarita Vega
- Department of Obstetrics and Gynecology, School of Medicine, University of Chile, Clinical Hospital, Santos Dumont # 999, Santiago, Chile.
| |
Collapse
|
30
|
McNamara KM, Moore NL, Hickey TE, Sasano H, Tilley WD. Complexities of androgen receptor signalling in breast cancer. Endocr Relat Cancer 2014; 21:T161-81. [PMID: 24951107 DOI: 10.1530/erc-14-0243] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
While the clinical benefit of androgen-based therapeutics in breast cancer has been known since the 1940s, we have only recently begun to fully understand the mechanisms of androgen action in breast cancer. Androgen signalling pathways can have either beneficial or deleterious effects in breast cancer depending on the breast cancer subtype and intracellular context. This review discusses our current knowledge of androgen signalling in breast cancer, including the relationship between serum androgens and breast cancer risk, the prognostic significance of androgen receptor (AR) expression in different breast cancer subtypes and the downstream molecular pathways mediating androgen action in breast cancer cells. Intracrine androgen metabolism has also been discussed and proposed as a potential mechanism that may explain some of the reported differences regarding dichotomous androgen actions in breast cancers. A better understanding of AR signalling in this disease is critical given the current resurgence in interest in utilising contemporary AR-directed therapies for breast cancer and the need for biomarkers that will accurately predict clinical response.
Collapse
Affiliation(s)
- Keely M McNamara
- Department of PathologyTohoku University School of Medicine, Miyagi, Sendai, JapanDame Roma Mitchell Cancer Research LaboratoriesDiscipline of Medicine, The University of Adelaide and Hanson Institute, DX 650801, Adelaide, South Australia 5005, Australia
| | - Nicole L Moore
- Department of PathologyTohoku University School of Medicine, Miyagi, Sendai, JapanDame Roma Mitchell Cancer Research LaboratoriesDiscipline of Medicine, The University of Adelaide and Hanson Institute, DX 650801, Adelaide, South Australia 5005, Australia
| | - Theresa E Hickey
- Department of PathologyTohoku University School of Medicine, Miyagi, Sendai, JapanDame Roma Mitchell Cancer Research LaboratoriesDiscipline of Medicine, The University of Adelaide and Hanson Institute, DX 650801, Adelaide, South Australia 5005, Australia
| | - Hironobu Sasano
- Department of PathologyTohoku University School of Medicine, Miyagi, Sendai, JapanDame Roma Mitchell Cancer Research LaboratoriesDiscipline of Medicine, The University of Adelaide and Hanson Institute, DX 650801, Adelaide, South Australia 5005, Australia
| | - Wayne D Tilley
- Department of PathologyTohoku University School of Medicine, Miyagi, Sendai, JapanDame Roma Mitchell Cancer Research LaboratoriesDiscipline of Medicine, The University of Adelaide and Hanson Institute, DX 650801, Adelaide, South Australia 5005, Australia
| |
Collapse
|
31
|
Campagnoli C, Berrino F, Venturelli E, Abbà C, Biglia N, Brucato T, Cogliati P, Danese S, Donadio M, Zito G, Pasanisi P. Metformin decreases circulating androgen and estrogen levels in nondiabetic women with breast cancer. Clin Breast Cancer 2014; 13:433-8. [PMID: 24267731 DOI: 10.1016/j.clbc.2013.08.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 07/08/2013] [Accepted: 08/26/2013] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Diabetic patients treated with metformin have a lower risk of developing BC or a better BC prognosis. Metformin might reduce cancer growth through direct antiproliferative effects or through indirect mechanisms, particularly the reduction of insulin. In a randomized study on nondiabetic BC patients in natural menopause with high testosterone levels, we observed a significant decrease in insulin and in testosterone levels with metformin 1500 mg/d compared with 1000 mg/d. We present the results of a new analysis of our study on the effect of metformin on the bioavailability of sex hormones. PATIENTS AND METHODS One hundred twenty-four eligible women were initially invited to take metformin 500 mg/d for 3 months. The 108 women who completed the first 3 months continued the study using 1000 mg/d for 1 month. The women were then randomized into 2 groups, and, for the subsequent 5 months, 1 group increased the dose to 1500 mg/d, and the other group continued with 1000 mg/d. RESULTS Ninety-six women completed the study, 43 receiving metformin 1500 mg/day, and 53 receiving 1000 mg/day. The women receiving 1500 mg/d showed a greater and significant reduction of free testosterone (-29%) and estradiol (-38%), a borderline significant reduction of estrone and insulin-like growth factor-1, and a nonsignificant reduction of androstenedione. They also showed a nonsignificant increase of dehydroepiandrosterone sulfate. CONCLUSION Metformin does not interfere with the production of dehydroepiandrosterone sulfate. Besides, it decreases estradiol levels, basically through the reduction of testosterone. These hormonal changes might have clinical relevance.
Collapse
Affiliation(s)
- Carlo Campagnoli
- Unit of Endocrinological Gynecology, Ospedale Sant'Anna di Torino, Turin, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hanamura T, Niwa T, Gohno T, Kurosumi M, Takei H, Yamaguchi Y, Ito KI, Hayashi SI. Possible role of the aromatase-independent steroid metabolism pathways in hormone responsive primary breast cancers. Breast Cancer Res Treat 2013; 143:69-80. [PMID: 24292869 DOI: 10.1007/s10549-013-2788-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 11/22/2013] [Indexed: 12/21/2022]
Abstract
Aromatase inhibitors (AIs) exert antiproliferative effects by reducing local estrogen production from androgens in postmenopausal women with hormone-responsive breast cancer. Previous reports have shown that androgen metabolites generated by the aromatase-independent enzymes, 5α-androstane-3β, 17β-diol (3β-diol), androst-5-ene-3β, and 17β-diol (A-diol), also activate estrogen receptor (ER) α. Estradiol (E2) can also reportedly be generated from estrone sulfate (E1S) pooled in the plasma. Estrogenic steroid-producing aromatase-independent pathways have thus been proposed as a mechanism of AI resistance. However, it is unclear whether these pathways are functional in clinical breast cancer. To investigate this issue, we assessed the transcriptional activities of ER in 45 ER-positive human breast cancers using the adenovirus estrogen-response element-green fluorescent protein assay and mRNA expression levels of the ER target gene, progesterone receptor, as indicators of ex vivo and in vivo ER activity, respectively. We also determined mRNA expression levels of 5α-reductase type 1 (SRD5A1) and 3β-hydroxysteroid dehydrogenase type 1 (3β-HSD type 1; HSD3B1), which produce 3β-diol from androgens, and of steroid sulfatase (STS) and 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD type 1; HSD17B1), which produce E2 or A-diol from E1S or dehydroepiandrosterone sulfate. SRD5A1 and HSD3B1 expression levels were positively correlated with ex vivo and in vivo ER activities. STS and HSD17B1 expression levels were positively correlated with in vivo ER activity alone. Elevated expression levels of these steroid-metabolizing enzymes in association with high in vivo ER activity were particularly notable in postmenopausal patients. Analysis of the expression levels of steroid-metabolizing enzymes revealed positive correlations between SRD5A1 and HSD3B1, and STS and HSD17B1. These findings suggest that the SRD5A1-HSD3B1 as well as the STS-HSD17B pathways, could contributes to ER activation, especially postmenopause. These pathways might function as an alternative estrogenic steroid-producing, aromatase-independent pathways.
Collapse
Affiliation(s)
- Toru Hanamura
- Department of Molecular and Functional Dynamics Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan,
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Chottanapund S, Van Duursen MBM, Navasumrit P, Hunsonti P, Timtavorn S, Ruchirawat M, Van den Berg M. Effect of androgens on different breast cancer cells co-cultured with or without breast adipose fibroblasts. J Steroid Biochem Mol Biol 2013; 138:54-62. [PMID: 23562642 DOI: 10.1016/j.jsbmb.2013.03.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 03/25/2013] [Accepted: 03/26/2013] [Indexed: 11/27/2022]
Abstract
About 70% of breast tumors express androgen receptors. In addition, there is clinical evidence suggesting that androgens can inhibit mammary epithelial proliferation. Vice versa, there is also significant evidence indicating that androgens can increase the risk of breast cancer via multiple mechanisms, e.g. direct conversion to estrogens that can bind to the estrogen receptor and thereby stimulate cell proliferation. We examined the effect of testosterone (T) and dihydroxytestosterone (DHT) on cell proliferation, pS2 and Ki-67 expression in three different breast cancer cell lines alone or in co-culture with primary human breast adipose fibroblasts (BAFs) obtained from breast cancer patients. In the co-cultures, T induced cell proliferation, pS2 and Ki-67 expression in the estrogen receptor positive (ER(+)) MCF-7 and T47D cells. This was not observed in the (ER(-)) MDA-MB-231 cells. The differences might be explained by the high expression of aromatase, which converts androgens to estrogens in BAFs followed by ER-mediated cell proliferation. In line with this absence of increased cell proliferation, pS2 and Ki-67 expression was observed in the presence of DHT, which is not a substrate for aromatase. In contrast, DHT caused a significant suppression of cell proliferation (68% and 38%), pS2 and Ki-67 expression in the (ER(+)) MCF-7 and T47D cells. More importantly, DHT decreased cell proliferation in (ER(-)) MDA-MB-231 cells by 38%. The results suggest that androgens that cannot be aromatized, like DHT, may provide a perspective for treatment of breast cancer patients, especially those with triple negative breast cancer.
Collapse
Affiliation(s)
- Suthat Chottanapund
- Division of Environmental Toxicology, Chulabhorn Graduate Institute, Bangkok, Thailand; Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, Thailand; Center of Excellence on Environmental Health, Toxicology and management of Chemicals, Bangkok, Thailand; Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands; Bamrasnaradura Infectious Diseases Institute, Ministry of Public Health, Thailand.
| | | | | | | | | | | | | |
Collapse
|
34
|
Androgen glucuronides analysis by liquid chromatography tandem-mass spectrometry: could it raise new perspectives in the diagnostic field of hormone-dependent malignancies? J Chromatogr B Analyt Technol Biomed Life Sci 2013; 940:24-34. [PMID: 24140653 DOI: 10.1016/j.jchromb.2013.09.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 08/09/2013] [Accepted: 09/18/2013] [Indexed: 01/14/2023]
Abstract
Breast and prostate constitute organs of intense steroidogenic activity. Clinical and epidemiologic data provide strong evidence on the influence of androgens and estrogens on the risk of typical hormone-dependent malignancies, like breast and prostate cancer. Recent studies have focused on the role of androgen metabolites in regulating androgen concentrations in hormone-sensitive tissues. Steroid glucuronidation has been suggested to have a prominent role in controlling the levels and the biological activity of unconjugated androgens. It is well-established that serum levels of androgen glucuronides reflect androgen metabolism in androgen-sensitive tissues. Quantitative analysis of androgen metabolites in blood specimens is the only minimally invasive approach permitting an accurate estimate of the total pool of androgens. During the past years, androgen glucuronides analysis most often involved radioimmunoassays (RIA) or direct immunoassays, both methods bearing serious limitations. However, recent impressive technical advances in mass spectrometry, and particularly in high performance liquid chromatography coupled with mass spectrometry (LC-MS/MS), have overcome these drawbacks enabling the simultaneous, quantitative analysis of multiple steroids even at low concentrations. Blood androgen profiling by LC-MS/MS, a robust and reliable technique of high selectivity, sensitivity, specificity, precision and accuracy emerges as a promising new approach in the study of human pathology. The present review offers a contemporary insight in androgen glucuronides profiling through the application of LC-MS/MS, highlighting new perspectives in the study of steroids and their implication in hormone-dependent malignancies.
Collapse
|
35
|
Postmenopausal breast cancer, androgens, and aromatase inhibitors. Breast Cancer Res Treat 2013; 139:1-11. [PMID: 23572296 DOI: 10.1007/s10549-013-2505-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 03/25/2013] [Indexed: 02/06/2023]
Abstract
Recent data can help to better define the long debated relationship between androgens and breast cancer (BC) after menopause. We reviewed the available literature data on: the origin of androgens after menopause, the association between circulating androgens and BC incidence and recurrence, the relationship between circulating and intratumoral hormones, the prognostic significance of the presence of androgen receptors (ARs) in the different BC subtypes, the androgen effect on BC cell lines, and the relationship between androgens and aromatase inhibitors. Epidemiological, clinical, and preclinical data on the role of androgens and of ARs on estrogen receptor (ER)-negative BC are somewhat controversial. However, most preclinical studies suggest that activated ARs, when present, have a proliferative effect, particularly in HER2 expressing cell lines, due to the cross-talk between AR and HER2 pathways. As regards ER-positive BC, epidemiological studies associate androgen levels with increased incidence and risk of recurrences, whilst clinical studies associate the AR positivity with a better prognosis. Preclinical studies suggest that the action of androgens is bidirectional: mainly proliferative, because circulating androgens are the precursors of estrogens, but also anti-proliferative, because AR activation restrains ER activity. The relative increase of androgenic action that follows the blocking of androgen aromatization into estrogens by aromatase inhibitors (AIs), could contribute to their therapeutic efficacy in AR-positive cases. Available data, although defining a complex picture, suggest that circulating androgen levels are clinically relevant, particularly when AIs are used.
Collapse
|