1
|
Tien AH, Sadar MD. Treatments Targeting the Androgen Receptor and Its Splice Variants in Breast Cancer. Int J Mol Sci 2024; 25:1817. [PMID: 38339092 PMCID: PMC10855698 DOI: 10.3390/ijms25031817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/15/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Breast cancer is a major cause of death worldwide. The complexity of endocrine regulation in breast cancer may allow the cancer cells to escape from a particular treatment and result in resistant and aggressive disease. These breast cancers usually have fewer treatment options. Targeted therapies for cancer patients may offer fewer adverse side effects because of specificity compared to conventional chemotherapy. Signaling pathways of nuclear receptors, such as the estrogen receptor (ER), have been intensively studied and used as therapeutic targets. Recently, the role of the androgen receptor (AR) in breast cancer is gaining greater attention as a therapeutic target and as a prognostic biomarker. The expression of constitutively active truncated AR splice variants in breast cancer is a possible mechanism contributing to treatment resistance. Therefore, targeting both the full-length AR and AR variants, either through the activation or suppression of AR function, depending on the status of the ER, progesterone receptor, or human epidermal growth factor receptor 2, may provide additional treatment options. Studies targeting AR in combination with other treatment strategies are ongoing in clinical trials. The determination of the status of nuclear receptors to classify and identify patient subgroups will facilitate optimized and targeted combination therapies.
Collapse
Affiliation(s)
- Amy H. Tien
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada
| | - Marianne D. Sadar
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| |
Collapse
|
2
|
Crooks TA, Madison JD, Walsh DM, Herbert WG, Jeraldo PR, Chia N, Cliby WA, Kaufmann SH, Walther-Antonio MRS. Porphyromonas somerae Invasion of Endometrial Cancer Cells. Front Microbiol 2021; 12:674835. [PMID: 34367083 PMCID: PMC8343132 DOI: 10.3389/fmicb.2021.674835] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/25/2021] [Indexed: 12/24/2022] Open
Abstract
Recent evidence suggests an association between endometrial cancer and the understudied bacterial species Porphyromonas somerae. This association was demonstrated in previous work that indicated a significantly enriched abundance of P. somerae in the uterine microbiome of endometrial cancer patients. Given the known associations of the Porphyromonas genus and oral cancer, we hypothesized that P. somerae may play a similar pathogenic role in endometrial cancer via intracellular activity. Before testing our hypothesis, we first characterized P. somerae biology, as current background data is limited. These novel characterizations include growth curves in liquid medium and susceptibility tests to antibiotics. We tested our hypothesis by examining growth changes in response to 17β-estradiol, a known risk factor for endometrial cancer, followed by metabolomic profiling in the presence and absence of 17β-estradiol. We found that P. somerae exhibits increased growth in the presence of 17β-estradiol of various concentrations. However, we did not find significant changes in metabolite levels in response to 17β-estradiol. To study direct host-microbe interactions, we used in vitro invasion assays under hypoxic conditions and found evidence for intracellular invasion of P. somerae in endometrial adenocarcinoma cells. We also examined these interactions in the presence of 17β-estradiol but did not observe changes in invasion frequency. Invasion was shown using three lines of evidence including visualization via differential staining and brightfield microscopy, increased frequency of bacterial recovery after co-culturing, and in silico methods to detail relevant genomic and transcriptomic components. These results underscore potential intracellular phenotypes of P. somerae within the uterine microbiome. Furthermore, these results raise new questions pertaining to the role of P. somerae in the progression of endometrial cancer.
Collapse
Affiliation(s)
- Taylor A Crooks
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Microbiology and Immunology, University of Minnesota Twin Cities, Minneapolis, MN, United States
| | - Joseph D Madison
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States.,Biology Department, University of Massachusetts Boston, Boston, MA, United States.,Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Dana M Walsh
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
| | - William G Herbert
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States.,Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Patricio R Jeraldo
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Nicholas Chia
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - William A Cliby
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, United States
| | - Scott H Kaufmann
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | - Marina R S Walther-Antonio
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Surgery, Mayo Clinic, Rochester, MN, United States.,Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
3
|
van Barele M, Heemskerk-Gerritsen BAM, Louwers YV, Vastbinder MB, Martens JWM, Hooning MJ, Jager A. Estrogens and Progestogens in Triple Negative Breast Cancer: Do They Harm? Cancers (Basel) 2021; 13:2506. [PMID: 34063736 PMCID: PMC8196589 DOI: 10.3390/cancers13112506] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/29/2021] [Accepted: 05/17/2021] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancers (TNBC) occur more frequently in younger women and do not express estrogen receptor (ER) nor progesterone receptor (PR), and are therefore often considered hormone-insensitive. Treatment of premenopausal TNBC patients almost always includes chemotherapy, which may lead to premature ovarian insufficiency (POI) and can severely impact quality of life. Hormone replacement therapy (HRT) is contraindicated for patients with a history of hormone-sensitive breast cancer, but the data on safety for TNBC patients is inconclusive, with a few randomized trials showing increased risk-ratios with wide confidence intervals for recurrence after HRT. Here, we review the literature on alternative pathways from the classical ER/PR. We find that for both estrogens and progestogens, potential alternatives exist for exerting their effects on TNBC, ranging from receptor conversion, to alternative receptors capable of binding estrogens, as well as paracrine pathways, such as RANK/RANKL, which can cause progestogens to indirectly stimulate growth and metastasis of TNBC. Finally, HRT may also influence other hormones, such as androgens, and their effects on TNBCs expressing androgen receptors (AR). Concluding, the assumption that TNBC is completely hormone-insensitive is incorrect. However, the direction of the effects of the alternative pathways is not always clear, and will need to be investigated further.
Collapse
Affiliation(s)
- Mark van Barele
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| | - Bernadette A. M. Heemskerk-Gerritsen
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| | - Yvonne V. Louwers
- Department of Obstetrics and Gynaecology, Erasmus MC, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands;
| | - Mijntje B. Vastbinder
- Department of Internal Medicine, Ijsselland Hospital, Prins Constantijnweg 2, 2906 ZC Capelle aan den IJssel, The Netherlands;
| | - John W. M. Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| | - Maartje J. Hooning
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (M.v.B.); (B.A.M.H.-G.); (J.W.M.M.); (M.J.H.)
| |
Collapse
|
4
|
Crooks TA, Madison JD, Walsh DM, Herbert WG, Jeraldo PR, Chia N, Cliby WA, Kaufmann SH, Walther-Antonio MRS. Porphyromonas somerae Invasion of Endometrial Cancer Cells. Front Microbiol 2021. [PMID: 34367083 DOI: 10.3389/fmicb.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023] Open
Abstract
Recent evidence suggests an association between endometrial cancer and the understudied bacterial species Porphyromonas somerae. This association was demonstrated in previous work that indicated a significantly enriched abundance of P. somerae in the uterine microbiome of endometrial cancer patients. Given the known associations of the Porphyromonas genus and oral cancer, we hypothesized that P. somerae may play a similar pathogenic role in endometrial cancer via intracellular activity. Before testing our hypothesis, we first characterized P. somerae biology, as current background data is limited. These novel characterizations include growth curves in liquid medium and susceptibility tests to antibiotics. We tested our hypothesis by examining growth changes in response to 17β-estradiol, a known risk factor for endometrial cancer, followed by metabolomic profiling in the presence and absence of 17β-estradiol. We found that P. somerae exhibits increased growth in the presence of 17β-estradiol of various concentrations. However, we did not find significant changes in metabolite levels in response to 17β-estradiol. To study direct host-microbe interactions, we used in vitro invasion assays under hypoxic conditions and found evidence for intracellular invasion of P. somerae in endometrial adenocarcinoma cells. We also examined these interactions in the presence of 17β-estradiol but did not observe changes in invasion frequency. Invasion was shown using three lines of evidence including visualization via differential staining and brightfield microscopy, increased frequency of bacterial recovery after co-culturing, and in silico methods to detail relevant genomic and transcriptomic components. These results underscore potential intracellular phenotypes of P. somerae within the uterine microbiome. Furthermore, these results raise new questions pertaining to the role of P. somerae in the progression of endometrial cancer.
Collapse
Affiliation(s)
- Taylor A Crooks
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Microbiology and Immunology, University of Minnesota Twin Cities, Minneapolis, MN, United States
| | - Joseph D Madison
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
- Biology Department, University of Massachusetts Boston, Boston, MA, United States
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Dana M Walsh
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
| | - William G Herbert
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, United States
| | - Patricio R Jeraldo
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - Nicholas Chia
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
| | - William A Cliby
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, United States
| | - Scott H Kaufmann
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | - Marina R S Walther-Antonio
- Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
5
|
Secreto G, Girombelli A, Krogh V. Androgen excess in breast cancer development: implications for prevention and treatment. Endocr Relat Cancer 2019; 26:R81-R94. [PMID: 30403656 DOI: 10.1530/erc-18-0429] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 11/05/2018] [Indexed: 12/15/2022]
Abstract
The aim of this review is to highlight the pivotal role of androgen excess in the development of breast cancer. Available evidence suggests that testosterone controls breast epithelial growth through a balanced interaction between its two active metabolites: cell proliferation is promoted by estradiol while it is inhibited by dihydrotestosterone. A chronic overproduction of testosterone (e.g. ovarian stromal hyperplasia) results in an increased estrogen production and cell proliferation that are no longer counterbalanced by dihydrotestosterone. This shift in the androgen/estrogen balance partakes in the genesis of ER-positive tumors. The mammary gland is a modified apocrine gland, a fact rarely considered in breast carcinogenesis. When stimulated by androgens, apocrine cells synthesize epidermal growth factor (EGF) that triggers the ErbB family receptors. These include the EGF receptor and the human epithelial growth factor 2, both well known for stimulating cellular proliferation. As a result, an excessive production of androgens is capable of directly stimulating growth in apocrine and apocrine-like tumors, a subset of ER-negative/AR-positive tumors. The key role of androgen excess in the genesis of different subtypes of breast cancer has significant clinical implications for both treatment and prevention. Our belief stems from a thorough analysis of the literature, where an abundance of evidence is present to justify a clinical trial that would investigate the effectiveness of treating the underlying excessive androgen production.
Collapse
Affiliation(s)
- Giorgio Secreto
- Epidemiology and Prevention Unit, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milano, Italy
| | - Alessandro Girombelli
- Anesthesia and Critical Care Medicine, ASST - Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Vittorio Krogh
- Epidemiology and Prevention Unit, Fondazione IRCCS - Istituto Nazionale dei Tumori, Milano, Italy
| |
Collapse
|
6
|
Pre-diagnostic sex hormone levels and survival among breast cancer patients. Breast Cancer Res Treat 2019; 174:749-758. [PMID: 30604001 DOI: 10.1007/s10549-018-05121-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/24/2018] [Indexed: 12/21/2022]
Abstract
PURPOSE Higher levels of circulating sex steroid hormones are associated with increased breast cancer risk, though their association with prognosis remains unclear. We evaluated the association between circulating sex hormone levels and breast cancer survival in two large cohorts. METHODS We evaluated this association among 2073 breast cancer cases from the Nurses' Health Study (NHS) and Nurses' Health Study II (NHSII) cohorts. Women in this analysis provided a blood sample in 1989-1990 (NHS) or in 1996-1999 (NHSII) and were subsequently diagnosed with breast cancer. Levels of estradiol (postmenopausal women only), testosterone, dehydroepiandrosterone-sulfate (DHEAS), and sex hormone-binding globulin (SHBG) were measured in plasma. Cox proportional hazards models were used to estimate hazard ratios (HR) and 95% confidence intervals (95% CI) for survival, adjusting for patient and tumor characteristics. RESULTS A total of 639 deaths and 160 breast cancer deaths occurred over follow-up through 2015. Compared to women in the lowest quartile, postmenopausal women in the highest quartile of estradiol experienced a 1.43-fold overall mortality rate (HR 1.43, 95% CI 1.03-1.97, P-trend = 0.04) and a nonsignificantly higher breast cancer mortality rate (HR 1.50, 95% CI 0.75-2.98, P-trend = 0.12). Higher DHEAS levels were nonsignificantly associated with better overall survival (HRQ4vsQ1=0.79, 95% CI 0.57-1.10, P-trend = 0.05), though not with breast cancer survival. No associations were observed between testosterone or SHBG and survival. CONCLUSIONS Pre-diagnostic postmenopausal circulating estradiol levels were modestly associated with worse survival among breast cancer patients. Further studies should evaluate whether circulating hormone levels at diagnosis predict cancer prognosis or treatment response.
Collapse
|
7
|
Kensler KH, Beca F, Baker GM, Heng YJ, Beck AH, Schnitt SJ, Hazra A, Rosner BA, Eliassen AH, Hankinson SE, Brown M, Tamimi RM. Androgen receptor expression in normal breast tissue and subsequent breast cancer risk. NPJ Breast Cancer 2018; 4:33. [PMID: 30276234 PMCID: PMC6155011 DOI: 10.1038/s41523-018-0085-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 01/09/2023] Open
Abstract
Sex steroid hormone signaling is critical in the development of breast cancers, although the role of the androgen receptor remains unclear. This study evaluated androgen receptor (AR) expression in normal breast tissue as a potential marker of breast cancer risk. We conducted a nested case-control study of women with benign breast disease (BBD) within the Nurses' Health Studies. Epithelial AR expression was assessed by immunohistochemistry in normal tissue from the BBD biopsy and the percent of positive nuclei was estimated in ordinal categories of 10% for 78 breast cancer cases and 276 controls. Logistic regression models adjusting for the matching factors and BBD lesion type were used to calculate odds ratios (ORs) for the association between AR expression (tertiles: ≤10%, 11-30%, and >30%) and breast cancer risk. AR expression in normal breast tissue was not associated with subsequent breast cancer risk (ORT3vsT1 = 0.9, 95% CI = 0.4-1.8, p trend = 0.68). In comparison with low AR/low ER women, ORs of 0.4 (95% CI = 0.1-1.2) for high AR/high ER women, 1.8 (95% CI = 0.4-7.8) for low AR/high ER women, and 0.7 (95% CI = 0.3-1.6) for high AR/low ER women were observed (p interaction = 0.21). Ki67 did not modify the association between AR expression and breast cancer risk (p interaction = 0.75). There was little evidence for an overall association between AR expression in normal breast tissue and breast cancer risk. These findings did not show that the AR association varied by Ki67 expression in normal breast tissue, though there was suggestive heterogeneity by ER expression.
Collapse
Affiliation(s)
- Kevin H Kensler
- 1Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215 USA.,2Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115 USA
| | - Francisco Beca
- 3Department of Pathology, Stanford University School of Medicine, Stanford, CA 94035 USA
| | - Gabrielle M Baker
- 4Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02215 USA.,5Harvard Medical School, Boston, MA 02215 USA
| | - Yujing J Heng
- 4Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02215 USA.,5Harvard Medical School, Boston, MA 02215 USA
| | | | - Stuart J Schnitt
- 5Harvard Medical School, Boston, MA 02215 USA.,7Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115 USA
| | - Aditi Hazra
- 5Harvard Medical School, Boston, MA 02215 USA.,8Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115 USA
| | - Bernard A Rosner
- 5Harvard Medical School, Boston, MA 02215 USA.,9Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115 USA.,10Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115 USA
| | - A Heather Eliassen
- 2Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115 USA.,9Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115 USA
| | - Susan E Hankinson
- 2Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115 USA.,5Harvard Medical School, Boston, MA 02215 USA.,9Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115 USA.,11Department of Biostatistics and Epidemiology, University of Massachusetts School of Public Health and Health Sciences, Amherst, MA 01003 USA
| | - Myles Brown
- 1Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215 USA.,5Harvard Medical School, Boston, MA 02215 USA
| | - Rulla M Tamimi
- 2Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115 USA.,9Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115 USA
| |
Collapse
|
8
|
Wasniewski T, Woclawek-Potocka I. Altered expression of lysophosphatidic acid receptors, in association with the synthesis of estrogens and androgens in type 1 endometrial cancer biology. Gynecol Endocrinol 2018; 34:422-427. [PMID: 29182028 DOI: 10.1080/09513590.2017.1409707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
To establish association between two main lysophosphatidic acid (LPA) receptors (LPAR2 and LPAR1) with the synthesis of estrogens and androgens in type-1 endometrial carcinoma (EC), we evaluated correlation of LPARs expression with expression of steroid 5 alpha-reductase 2 - aromatase (SRD5A2), or cytochrome P450 family 19 subfamily A member 1-5α-reductase (CYP19A1) in EC. Moreover, we aimed to investigate SRD5A2 and CYP19A1 expression in type 1 endometrial cancer and normal endometrium with its correlation to selected clinicopathological features. The studied cancerous samples showed higher CYP19A1 and SRD5A2 expression comparing to normal endometria. We also documented positive correlations between LPAR1 and LPAR2 with responsible for proliferation SRD5A2 in EC tissue which suggests that intratumoral estrogen metabolism and synthesis are pivotal in endometrial carcinoma progression, with the involvement of LPA in this process. However, positive correlation between CYP19A1 and LPAR1 accounts for supporting role of LPA acting via LPAR1 in intratumoral DHT concentration and the ethiology of endometrial cancer progression. Also, owing to the highest expression of LPARs, CYP19A1 and SRD5A2 as well as their association with depth of myoinvasion and FIGO stage LPAR2 and LPAR1 seem to be the efficient candidate prognostic markers in the individual, targeted therapies for EC.
Collapse
Affiliation(s)
- Tomasz Wasniewski
- a Department of Gynecology and Obstetrics, Faculty of Medical Sciences , University of Warmia and Masuria , Olsztyn , Poland
| | - Izabela Woclawek-Potocka
- b Department of Gamete and Embryo Biology , Institute of Animal Reproduction and Food Research, Polish Academy of Sciences , Olsztyn , Poland
| |
Collapse
|
9
|
Khatun A, Shimozawa M, Kito H, Kawaguchi M, Fujimoto M, Ri M, Kajikuri J, Niwa S, Fujii M, Ohya S. Transcriptional Repression and Protein Degradation of the Ca 2+-Activated K + Channel K Ca1.1 by Androgen Receptor Inhibition in Human Breast Cancer Cells. Front Physiol 2018; 9:312. [PMID: 29713287 PMCID: PMC5911984 DOI: 10.3389/fphys.2018.00312] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/14/2018] [Indexed: 01/14/2023] Open
Abstract
The large-conductance Ca2+-activated K+ channel KCa1.1 plays an important role in the promotion of breast cancer cell proliferation and metastasis. The androgen receptor (AR) is proposed as a therapeutic target for AR-positive advanced triple-negative breast cancer. We herein investigated the effects of a treatment with antiandrogens on the functional activity, activation kinetics, transcriptional expression, and protein degradation of KCa1.1 in human breast cancer MDA-MB-453 cells using real-time PCR, Western blotting, voltage-sensitive dye imaging, and whole-cell patch clamp recording. A treatment with the antiandrogen bicalutamide or enzalutamide for 48 h significantly suppressed (1) depolarization responses induced by paxilline (PAX), a specific KCa1.1 blocker and (2) PAX-sensitive outward currents induced by the depolarizing voltage step. The expression levels of KCa1.1 transcripts and proteins were significantly decreased in MDA-MB-453 cells, and the protein degradation of KCa1.1 mainly contributed to reductions in KCa1.1 activity. Among the eight regulatory β and γ subunits, LRRC26 alone was expressed at high levels in MDA-MB-453 cells and primary and metastatic breast cancer tissues, whereas no significant changes were observed in the expression levels of LRRC26 and activation kinetics of PAX-sensitive outward currents in MDA-MB-453 cells by the treatment with antiandrogens. The treatment with antiandrogens up-regulated the expression of the ubiquitin E3 ligases, FBW7, MDM2, and MDM4 in MDA-MB-453 cells, and the protein degradation of KCa1.1 was significantly inhibited by the respective siRNA-mediated blockade of FBW7 and MDM2. Based on these results, we concluded that KCa1.1 is an androgen-responsive gene in AR-positive breast cancer cells, and its down-regulation through enhancements in its protein degradation by FBW7 and/or MDM2 may contribute, at least in part, to the antiproliferative and antimetastatic effects of antiandrogens in breast cancer cells.
Collapse
Affiliation(s)
- Anowara Khatun
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Motoki Shimozawa
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Hiroaki Kito
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Mayu Kawaguchi
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Mayu Fujimoto
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Moe Ri
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Junko Kajikuri
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Satomi Niwa
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Masanori Fujii
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Susumu Ohya
- Division of Pathological Sciences, Department of Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan.,Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
10
|
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
|
11
|
Narayanan R, Dalton JT. Androgen Receptor: A Complex Therapeutic Target for Breast Cancer. Cancers (Basel) 2016; 8:cancers8120108. [PMID: 27918430 PMCID: PMC5187506 DOI: 10.3390/cancers8120108] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/01/2016] [Accepted: 11/23/2016] [Indexed: 12/29/2022] Open
Abstract
Molecular and histopathological profiling have classified breast cancer into multiple sub-types empowering precision treatment. Although estrogen receptor (ER) and human epidermal growth factor receptor (HER2) are the mainstay therapeutic targets in breast cancer, the androgen receptor (AR) is evolving as a molecular target for cancers that have developed resistance to conventional treatments. The high expression of AR in breast cancer and recent discovery and development of new nonsteroidal drugs targeting the AR provide a strong rationale for exploring it again as a therapeutic target in this disease. Ironically, both nonsteroidal agonists and antagonists for the AR are undergoing clinical trials, making AR a complicated target to understand in breast cancer. This review provides a detailed account of AR’s therapeutic role in breast cancer.
Collapse
Affiliation(s)
- Ramesh Narayanan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA.
| | - James T Dalton
- College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
12
|
D'Amato NC, Gordon MA, Babbs B, Spoelstra NS, Carson Butterfield KT, Torkko KC, Phan VT, Barton VN, Rogers TJ, Sartorius CA, Elias A, Gertz J, Jacobsen BM, Richer JK. Cooperative Dynamics of AR and ER Activity in Breast Cancer. Mol Cancer Res 2016; 14:1054-1067. [PMID: 27565181 DOI: 10.1158/1541-7786.mcr-16-0167] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/29/2016] [Accepted: 08/02/2016] [Indexed: 11/16/2022]
Abstract
Androgen receptor (AR) is expressed in 90% of estrogen receptor alpha-positive (ER+) breast tumors, but its role in tumor growth and progression remains controversial. Use of two anti-androgens that inhibit AR nuclear localization, enzalutamide and MJC13, revealed that AR is required for maximum ER genomic binding. Here, a novel global examination of AR chromatin binding found that estradiol induced AR binding at unique sites compared with dihydrotestosterone (DHT). Estradiol-induced AR-binding sites were enriched for estrogen response elements and had significant overlap with ER-binding sites. Furthermore, AR inhibition reduced baseline and estradiol-mediated proliferation in multiple ER+/AR+ breast cancer cell lines, and synergized with tamoxifen and fulvestrant. In vivo, enzalutamide significantly reduced viability of tamoxifen-resistant MCF7 xenograft tumors and an ER+/AR+ patient-derived model. Enzalutamide also reduced metastatic burden following cardiac injection. Finally, in a comparison of ER+/AR+ primary tumors versus patient-matched local recurrences or distant metastases, AR expression was often maintained even when ER was reduced or absent. These data provide preclinical evidence that anti-androgens that inhibit AR nuclear localization affect both AR and ER, and are effective in combination with current breast cancer therapies. In addition, single-agent efficacy may be possible in tumors resistant to traditional endocrine therapy, as clinical specimens of recurrent disease demonstrate AR expression in tumors with absent or refractory ER. IMPLICATIONS This study suggests that AR plays a previously unrecognized role in supporting E2-mediated ER activity in ER+/AR+ breast cancer cells, and that enzalutamide may be an effective therapeutic in ER+/AR+ breast cancers. Mol Cancer Res; 14(11); 1054-67. ©2016 AACR.
Collapse
Affiliation(s)
- Nicholas C D'Amato
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Michael A Gordon
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Beatrice Babbs
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Nicole S Spoelstra
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Kathleen C Torkko
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Valerie N Barton
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Thomas J Rogers
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Carol A Sartorius
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anthony Elias
- Department of Medicine, Division of Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jason Gertz
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Britta M Jacobsen
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
13
|
Novel targets for paclitaxel nano formulations: Hopes and hypes in triple negative breast cancer. Pharmacol Res 2016; 111:577-591. [PMID: 27461138 DOI: 10.1016/j.phrs.2016.07.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 12/30/2022]
Abstract
Triple negative breast cancer is defined as one of the utmost prevailing breast cancers worldwide, possessing an inadequate prognosis and treatment option limited to chemotherapy and radiotherapy, creating a challenge for researchers as far as developing a specific targeted therapy is concerned. The past research era has shown several promising outcomes for TNBC such as nano-formulations of the chemotherapeutic agents already used for the management of the malignant tumor. Taking a glance at paclitaxel nano formulations, it has been proven beneficial in several researches in the past decade; nevertheless its solubility is often a challenge to scientists in achieving success. We have henceforth discussed the basic heterogeneity of triple negative breast cancer along with the current management options as well as a brief outlook on pros and cons of paclitaxel, known as the most widely used chemotherapeutic agent for the treatment of the disease. We further analyzed the need of nanotechnology pertaining to the problems encountered with the current paclitaxel formulations available discussing the strategic progress in various nano-formulations till date taking into account the basic research strategies required in terms of solubility, permeability, physicochemical properties, active and passive targeting. A thorough review in recent advances in active targeting for TNBC was carried out whereby the various ligands which are at present finding its way into TNBC research such as hyaluronic acid, folic acid, transferrin, etc. were discussed. These ligands have specific receptor affinity to TNBC tumor cells hence can be beneficial for novel drug targeting approaches. Conversely, there are currently several novel strategies in the research pipeline whose targeting ligands have not yet been studied. Therefore, we reviewed upon the numerous novel receptor targets along with the respective nano-formulation aspects which have not yet been fully researched upon and could be exemplified as outstanding target strategies for TNBC which is currently an urgent requirement.
Collapse
|
14
|
Ito K, Miki Y, Suzuki T, McNamara KM, Sasano H. In situ androgen and estrogen biosynthesis in endometrial cancer: focus on androgen actions and intratumoral production. Endocr Relat Cancer 2016; 23:R323-35. [PMID: 27287451 DOI: 10.1530/erc-15-0470] [Citation(s) in RCA: 18] [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: 05/17/2016] [Accepted: 06/10/2016] [Indexed: 01/20/2023]
Abstract
In situ estrogen biosynthesis is considered to play pivotal roles in the development and progression of human endometrial carcinoma. However, the biological roles of androgen have remained virtually unknown. Various epidemiological studies have revealed that elevated serum androgen levels are generally associated with an increased risk of developing endometrial carcinoma; however, studies directly examining androgens in carcinoma tissues are relatively rare and reviews summarizing this information are scarce. Therefore, we summarized recent studies on androgens in endometrial carcinoma, especially focusing androgen actions and in situ androgen biosynthesis. Among the enzymes required for local biosynthesis of androgen, 17β-hydroxysteroid dehydrogenase type 5 (conversion from androstenedione to testosterone) and 5α-reductase (reduction of testosterone to dihydrotestosterone (DHT)) are the principal enzymes involved in the formation of biologically most potent androgen, DHT. Both enzymes and androgen receptor were expressed in endometrial carcinoma tissues, and in situ production of DHT has been reported to exist in endometrial carcinoma tissues. However, testosterone is not only a precursor of DHT production, but also a precursor of estradiol synthesis, as a substrate of the aromatase enzyme. Therefore, aromatase could be another key enzyme serving as a negative regulator for in situ production of DHT by reducing amounts of the precursor. In an in vitro study, DHT was reported to exert antiproliferative effects on endometrial carcinoma cells. Intracrine mechanisms of androgens, the downstream signals of AR, which are directly related to anticancer progression, and the clinical significance of DHT-AR pathway in the patients with endometrial carcinoma have, however, not been fully elucidated.
Collapse
Affiliation(s)
- Kiyoshi Ito
- Department of Disaster Obstetrics and GynecologyInternational Research Institute of Disaster Science (IRIDeS), Tohoku University, Sendai, Japan Department of Disaster Obstetrics and GynecologyTohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Miki
- Department of Disaster Obstetrics and GynecologyInternational Research Institute of Disaster Science (IRIDeS), Tohoku University, Sendai, Japan
| | - Takashi Suzuki
- Department of Pathology and HistotechnologyTohoku University Graduate School of Medicine, Sendai, Japan
| | - Keely May McNamara
- Department of PathologyTohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of PathologyTohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
15
|
Kumar P, Aggarwal R. An overview of triple-negative breast cancer. Arch Gynecol Obstet 2015; 293:247-69. [PMID: 26341644 DOI: 10.1007/s00404-015-3859-y] [Citation(s) in RCA: 457] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 08/18/2015] [Indexed: 12/22/2022]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is a heterogeneous group of tumors comprising various breast cancers simply defined by the absence of estrogen receptor, progesterone receptor and overexpression of human epidermal growth factor receptor 2 gene. In this review, we discuss the epidemiology, risk factors, clinical characteristics and prognostic variables of TNBC, and present the summary of recommended treatment strategies and all other available treatment options. METHODS We performed a systematic literature search using Medline and selected those articles which seemed relevant for this review. In addition, the ClinicalTrials.gov was also scanned for ongoing trials. RESULTS TNBC accounts for 10-20 % of all invasive breast cancers and has been found to be associated with African-American race, younger age, higher grade and mitotic index, and more advanced stage at diagnosis. Locoregional treatment is similar to other invasive breast cancer subtypes and involves surgery-mastectomy with or without adjuvant radiotherapy or breast conservation followed by adjuvant radiotherapy. Due to lack of drug-targetable receptors, chemotherapy is the only recommended systemic treatment to improve disease outcome. TNBC is sensitive to chemotherapy as demonstrated by high pathological complete response rates achieved after neoadjuvant chemotherapy, and this approach also allows for breast-conserving surgery. The peak risk of relapse is at 3 years after surgery, thereafter recurrence risk rapidly decreases. Survival after metastatic relapse is shorter as compared to other breast cancer subtypes, treatment options are few and response rates are poor and lack durability. Important molecular characteristics have now been identified that can subdivide this group of breast cancers further and can provide alternative systemic therapies. CONCLUSIONS To improve therapeutic outcome of TNBC, reliable predictive biomarkers and newer drugs against the known molecular pathways are required.
Collapse
Affiliation(s)
- Pankaj Kumar
- Department of Radiation Oncology, Max Super Speciality Hospital, Phase-6, Mohali, 160055, Punjab, India.
| | - Rupali Aggarwal
- Department of Radiation Oncology, Indus Super Speciality Hospital, Phase-1, Mohali, 160055, Punjab, India
| |
Collapse
|
16
|
Barton VN, D'Amato NC, Gordon MA, Lind HT, Spoelstra NS, Babbs BL, Heinz RE, Elias A, Jedlicka P, Jacobsen BM, Richer JK. Multiple molecular subtypes of triple-negative breast cancer critically rely on androgen receptor and respond to enzalutamide in vivo. Mol Cancer Ther 2015; 14:769-78. [PMID: 25713333 DOI: 10.1158/1535-7163.mct-14-0926] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 01/11/2015] [Indexed: 12/31/2022]
Abstract
Triple-negative breast cancer (TNBC) has the lowest 5-year survival rate of invasive breast carcinomas, and currently there are no approved targeted therapies for this aggressive form of the disease. The androgen receptor (AR) is expressed in up to one third of TNBC and we find that all AR(+) TNBC primary tumors tested display nuclear localization of AR, indicative of transcriptionally active receptors. While AR is most abundant in the "luminal AR (LAR)" molecular subtype of TNBC, here, for the first time, we use both the new-generation anti-androgen enzalutamide and AR knockdown to demonstrate that the other non-LAR molecular subtypes of TNBC are critically dependent on AR protein. Indeed, AR inhibition significantly reduces baseline proliferation, anchorage-independent growth, migration, and invasion and increases apoptosis in four TNBC lines (SUM159PT, HCC1806, BT549, and MDA-MB-231), representing three non-LAR TNBC molecular subtypes (mesenchymal-like, mesenchymal stem-like, and basal-like 2). In vivo, enzalutamide significantly decreases viability of SUM159PT and HCC1806 xenografts. Furthermore, mechanistic analysis reveals that AR activation upregulates secretion of the EGFR ligand amphiregulin (AREG), an effect abrogated by enzalutamide in vitro and in vivo. Exogenous AREG partially rescues the effects of AR knockdown on proliferation, migration, and invasion, demonstrating that upregulation of AREG is one mechanism by which AR influences tumorigenicity. Together, our findings indicate that non-LAR subtypes of TNBC are AR dependent and, moreover, that enzalutamide is a promising targeted therapy for multiple molecular subtypes of AR(+) TNBC.
Collapse
Affiliation(s)
- Valerie N Barton
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Nicholas C D'Amato
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Michael A Gordon
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Hanne T Lind
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Nicole S Spoelstra
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Beatrice L Babbs
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Richard E Heinz
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anthony Elias
- Department of Medicine, Division of Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Paul Jedlicka
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Britta M Jacobsen
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
17
|
Yoda T, McNamara KM, Miki Y, Takagi M, Rai Y, Ohi Y, Sagara Y, Tamaki K, Hirakawa H, Ishida T, Suzuki T, Ohuchi N, Sasano H. Intratumoral androgen metabolism and actions in invasive lobular carcinoma of the breast. Cancer Sci 2014; 105:1503-9. [PMID: 25230018 PMCID: PMC4462384 DOI: 10.1111/cas.12535] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/30/2014] [Accepted: 09/09/2014] [Indexed: 12/19/2022] Open
Abstract
Invasive lobular carcinoma (ILC) accounts for approximately 10% of all breast carcinomas and is characterized by higher levels of androgen receptor (AR) compared to invasive ductal carcinoma (IDC). Despite this potentially androgen-responsive environment, the combined importance of AR and androgen metabolism in non-neoplastic lobules and lobular carcinoma remains unknown. Therefore, in this study, we evaluated the status of pivotal androgen-producing enzymes 17β-hydroxysteroid dehydrogenase type 5 (17βHSD5) and 5α-reductase type 1 (5αRed1) in 178 cases of ILC and surrounding histologically non-neoplastic lobular tissue using immunohistochemistry. Androgen receptor prevalence was higher but androgenic enzymes lower in ILC than non-neoplastic lobules. In ILC cases the status of 5αRed1 and 17βHSD5 was inversely correlated with tumor size (P = 0.0053) and nuclear grade (P = 0.0290), and significantly associated with better overall survival of the patients (P = 0.0059). Based on these findings, we hypothesized that androgen signaling could act as a tumor suppressor. As previous studies suggested that androgens might partially act by increasing levels of the estrogen inactivating enzyme 17β-hydroxysteroid dehydrogenase type 2 (17βHSD2) in IDC tissues, this was reasonably considered a potential mechanism of androgen actions. Significantly positive correlation was detected between the status of androgenic enzymes and 17βHSD2 (P < 0.0001) and intratumoral 17βHSD2 was inversely correlated with tumor size in ILC (P = 0.0075). These correlations suggest one protective mode of androgen action could be through modulation of estrogen metabolism. Results of our present study indicated that androgen-producing enzymes could play pivotal protective roles in AR-enriched ILC cases.
Collapse
Affiliation(s)
- Tomomi Yoda
- Department of Pathology, Tohoku University Graduate School of MedicineSendai
| | - Keely May McNamara
- Department of Pathology, Tohoku University Graduate School of MedicineSendai
| | - Yasuhiro Miki
- Department of Pathology, Tohoku University Graduate School of MedicineSendai
| | - Mayu Takagi
- Department of Pathology, Tohoku University Graduate School of MedicineSendai
- Department of Surgery, Tohoku University Graduate School of MedicineSendai, Japan
| | - Yoshiaki Rai
- Department of Sagara Hospital, Social Medical Corporation HakuaikaiKagoshima, Japan
| | - Yasuyo Ohi
- Department of Sagara Hospital, Social Medical Corporation HakuaikaiKagoshima, Japan
| | - Yasuaki Sagara
- Department of Sagara Hospital, Social Medical Corporation HakuaikaiKagoshima, Japan
| | - Kentaro Tamaki
- Department of Pathology, Tohoku University Graduate School of MedicineSendai
- Department of Surgery, Tohoku University Graduate School of MedicineSendai, Japan
- Department of Nahanishi ClinicNaha, Japan
| | | | - Takanori Ishida
- Department of Surgery, Tohoku University Graduate School of MedicineSendai, Japan
| | - Takashi Suzuki
- Department of Pathology, Tohoku University Graduate School of MedicineSendai
| | - Noriaki Ohuchi
- Department of Surgery, Tohoku University Graduate School of MedicineSendai, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of MedicineSendai
| |
Collapse
|
18
|
Cochrane DR, Bernales S, Jacobsen BM, Cittelly DM, Howe EN, D'Amato NC, Spoelstra NS, Edgerton SM, Jean A, Guerrero J, Gómez F, Medicherla S, Alfaro IE, McCullagh E, Jedlicka P, Torkko KC, Thor AD, Elias AD, Protter AA, Richer JK. Role of the androgen receptor in breast cancer and preclinical analysis of enzalutamide. Breast Cancer Res 2014; 16:R7. [PMID: 24451109 PMCID: PMC3978822 DOI: 10.1186/bcr3599] [Citation(s) in RCA: 290] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 01/08/2014] [Indexed: 01/11/2023] Open
Abstract
Introduction The androgen receptor (AR) is widely expressed in breast cancers and has been proposed as a therapeutic target in estrogen receptor alpha (ER) negative breast cancers that retain AR. However, controversy exists regarding the role of AR, particularly in ER + tumors. Enzalutamide, an AR inhibitor that impairs nuclear localization of AR, was used to elucidate the role of AR in preclinical models of ER positive and negative breast cancer. Methods We examined nuclear AR to ER protein ratios in primary breast cancers in relation to response to endocrine therapy. The effects of AR inhibition with enzalutamide were examined in vitro and in preclinical models of ER positive and negative breast cancer that express AR. Results In a cohort of 192 women with ER + breast cancers, a high ratio of AR:ER (≥2.0) indicated an over four fold increased risk for failure while on tamoxifen (HR = 4.43). The AR:ER ratio had an independent effect on risk for failure above ER % staining alone. AR:ER ratio is also an independent predictor of disease-free survival (HR = 4.04, 95% CI: 1.68, 9.69; p = 0.002) and disease specific survival (HR = 2.75, 95% CI: 1.11, 6.86; p = 0.03). Both enzalutamide and bicalutamide inhibited 5-alpha-dihydrotestosterone (DHT)-mediated proliferation of breast cancer lines in vitro; however, enzalutamide uniquely inhibited estradiol (E2)-mediated proliferation of ER+/AR + breast cancer cells. In MCF7 xenografts (ER+/AR+) enzalutamide inhibited E2-driven tumor growth as effectively as tamoxifen by decreasing proliferation. Enzalutamide also inhibited DHT- driven tumor growth in both ER positive (MCF7) and negative (MDA-MB-453) xenografts, but did so by increasing apoptosis. Conclusions AR to ER ratio may influence breast cancer response to traditional endocrine therapy. Enzalutamide elicits different effects on E2-mediated breast cancer cell proliferation than bicalutamide. This preclinical study supports the initiation of clinical studies evaluating enzalutamide for treatment of AR+ tumors regardless of ER status, since it blocks both androgen- and estrogen- mediated tumor growth.
Collapse
|
19
|
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
|
20
|
Abstract
Dihydrotestosterone (DHT) is the most potent natural androgen in humans. There has been an increasing interest in this androgen and its role in the development of primary and secondary sexual characteristics as well as its potential roles in diseases ranging from prostate and breast cancer to Alzheimer's disease. Despite the range of pathologies shown to involve DHT there is little evidence for measurement of serum DHT in the management of these diseases. In this review we describe the physiology of DHT production and action, summarize current concepts in the role of DHT in the pathogenesis of various disorders of sexual development, compare current methods for the measurement of DHT and conclude on the clinical utility of DHT measurement. The clinical indications for the measurement of DHT in serum are: investigation of 5α reductase deficiency in infants with ambiguous genitalia and palpable gonads; men with delayed puberty and/or undescended testes; and to confirm the presence of active testicular tissue. Investigation is aided by the use of human chorionic gonadotrophin stimulation. Due to paucity of published data on this procedure, it is important to follow guidelines prescribed by the laboratory performing the analysis to ensure accurate interpretation.
Collapse
Affiliation(s)
- Paula M Marchetti
- SAS Steroid Centre, St James' University Hospital, Block 46, Leeds LS9 7TF, UK.
| | | |
Collapse
|
21
|
Secreto G, Venturelli E, Meneghini E, Carcangiu ML, Paolini B, Agresti R, Pellitteri C, Berrino F, Gion M, Cogliati P, Saragò G, Micheli A. Androgen receptors and serum testosterone levels identify different subsets of postmenopausal breast cancers. BMC Cancer 2012; 12:599. [PMID: 23241075 PMCID: PMC3554552 DOI: 10.1186/1471-2407-12-599] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 11/26/2012] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Androgen receptors (AR) are frequently expressed in breast cancers, but their implication in cancer growth is still controversial. In the present study, we further investigated the role of the androgen/AR pathway in breast cancer development. METHODS AR expression was evaluated by immunochemistry in a cohort of 528 postmenopausal breast cancer patients previously examined for the association of serum testosterone levels with patient and tumor characteristics. AR expression was classified according to the percentage of stained cells: AR-absent (0%) and AR-poorly (1%-30%), AR-moderately (>30%-60%), and AR-highly (>60%) positive. RESULTS Statistical analysis was performed in 451 patients who experienced natural menopause. AR-high expression was significantly related with low histologic grade and estrogen receptor (ER)- and progesterone receptor (PR)-positive status (P trend<0.001). Mean testosterone levels were significantly higher in the AR-high category than in the other categories combined (P=0.022), although a trend across the AR expression categories was not present. When women defined by ER status were analyzed separately, regression analysis in the ER-positive group showed a significant association of high testosterone levels with AR-highly-positive expression (OR 1.86; 95% CI, 1.10-3.16), but the association was essentially due to patients greater than or equal to 65 years (OR 2.42; 95% CI, 1.22-4.82). In ER-positive group, elevated testosterone levels appeared also associated with AR-absent expression, although the small number of patients in this category limited the appearance of significant effects (OR 1.92; 95% CI, 0.73-5.02): the association was present in both age groups (<65 and ≥65 years). In the ER-negative group, elevated testosterone levels were found associated (borderline significance) with AR-absent expression (OR 2.82, 95% CI, 0.98-8.06). In this ER-negative/AR-absent subset of tumors, elevated testosterone levels cannot stimulate cancer growth either directly or after conversion into estrogens, but they probably induce increased synthesis of some other substance that is responsible for cancer growth through binding to its specific receptor. CONCLUSIONS The findings in the present study confirm that testosterone levels are a marker of hormone-dependent breast cancer and suggest that the contemporary evaluation of ER status, AR expression, and circulating testosterone levels may identify different subsets of cancers whose growth may be influenced by androgens.
Collapse
Affiliation(s)
- Giorgio Secreto
- Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
|
23
|
Circulating sex hormones and tumor characteristics in postmenopausal breast cancer patients. A cross-sectional study. Int J Biol Markers 2012; 26:241-6. [PMID: 22180173 DOI: 10.5301/jbm.2011.8883] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2011] [Indexed: 01/09/2023]
Abstract
BACKGROUND To further investigate the role of sex hormones in breast cancer, we assessed the relations of circulating estradiol and testosterone to tumor size and estrogen receptor (ER) status. METHOD This was a cross-sectional study including 492 postmenopausal breast cancer patients. The relation of circulating hormones to patient and tumor characteristics was assessed using the Fisher or Cuzick tests. Multivariable logistic regression was used to estimate the odds ratios (ORs) of having tumors =2 cm (vs and <2 cm) and having ER-positive tumors (vs ER-negative) with increasing quartiles of estradiol and testosterone. RESULTS Mean estradiol and testosterone levels increased significantly with increasing tumor size. The ORs of tumors =2 cm increased significantly with increasing quartiles of estradiol (Ptrend and <0.001) and testosterone (Ptrend=0.005). When adjusted for estradiol, the association between testosterone and tumor size was no longer significant. Mean testosterone levels were higher in ER-positive than ER-negative patients (p and <0.001), while mean estradiol levels did not differ significantly between the two ER categories (p=0.192). The ORs of having an ER-positive tumor increased significantly with increasing quartiles of testosterone (Ptrend=0.002), whereas the increase with increasing estradiol quartiles was not significant (Ptrend=0.07). CONCLUSION The association of both hormones with tumor size implies that both are involved in tumor growth, testosterone mainly by conversion to estradiol. The strong association of testosterone with ER contrasts with the weak association of estradiol with ER and confirms testosterone as a marker of hormone-dependent tumors. These findings suggest that testosterone evaluation might be useful to better identify patients with hormone-dependent disease.
Collapse
|
24
|
Estrogens in the breast tissue: a systematic review. Cancer Causes Control 2011; 22:529-40. [PMID: 21286801 DOI: 10.1007/s10552-011-9729-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 01/17/2011] [Indexed: 01/24/2023]
Abstract
The role of estrogens in breast carcinogenesis has been investigated at the level of whole body (plasma) and cell (molecular, receptors, etc.). Growing attention focused on the breast tissue being an intracrine organ, with potentially important local estrogen production in the breast. However, very little is known about the local breast tissue estrogen levels. Understanding the role of the tissue estrogens in breast carcinogenesis might open new avenues in breast cancer prevention. This systematic review summarizes published studies that measured local estrogen levels in the breast and offers suggestions for strategies to fill gaps in our existing scientific knowledge.
Collapse
|
25
|
Zhao G, Lu L, Luu-The V, Fan Z, Labrie F, Pelletier G. Expression of 5α-reductase type 1 in breast cancer and adjacent non-malignant tissue: an immunohistochemical study. Horm Mol Biol Clin Investig 2010; 3:411-5. [PMID: 25961212 DOI: 10.1515/hmbci.2010.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Accepted: 09/08/2010] [Indexed: 11/15/2022]
Abstract
Intratumoral biosynthesis of sex steroids is thought to play a role in the pathogenesis and development of human breast cancer. There is evidence that androgens can inhibit the development and progression of breast cancer. Among the enzymes involved in the biosynthesis of androgens, 5α-reductase plays a key role by reducing testosterone to dihydrotestosterone, the most potent androgen. Two isoforms of 5α-reductase have been characterized and 5α-reductase type 1 is predominant in breast cancer tissue. We developed specific antibodies to 5α-reductase type 1 and studied the expression of the enzyme in 84 specimens of breast carcinoma and adjacent non-malignant tissues by immunohistochemistry. The results were correlated with the expression of androgen receptor, estrogen receptor α, progesterone receptor and CDC47, a cell division marker as well as the tumor stage, tumor size, nodal status and menopausal status. The expression of 5α-reductase type 1 in 61% of breast cancer specimens appeared significantly lower than that observed in normal adjacent tissues (87% of cases being positive). There was no significant correlation between 5α-reductase type 1 expression and the clinicopathological parameters studied. The decrease in 5α-reductase type 1 expression in breast cancer as compared to that observed in the adjacent normal tissues could play a role in the development and/or progression of the cancer by modifying the intratumoral levels of androgens.
Collapse
|
26
|
Suzuki T, Miki Y, Takagi K, Hirakawa H, Moriya T, Ohuchi N, Sasano H. Androgens in human breast carcinoma. Med Mol Morphol 2010; 43:75-81. [DOI: 10.1007/s00795-010-0494-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 01/06/2010] [Indexed: 12/14/2022]
|
27
|
Pelekanou V, Notas G, Sanidas E, Tsapis A, Castanas E, Kampa M. Testosterone membrane-initiated action in breast cancer cells: Interaction with the androgen signaling pathway and EPOR. Mol Oncol 2010; 4:135-49. [PMID: 20189893 PMCID: PMC5527892 DOI: 10.1016/j.molonc.2010.01.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 01/21/2010] [Accepted: 01/22/2010] [Indexed: 12/29/2022] Open
Abstract
Membrane-initiated androgen actions have now been acknowledged, even though a specific binding site has not been biochemically characterized yet. Recent data indicate that testosterone-BSA, a non-permeable testosterone analog, can exert specific actions in breast cancer cell lines, including proper transcriptional effects, independent of the intracellular androgen sites. In the present work we explore the effects of testosterone-BSA in two specifically modified pathways, revealed by early trascriptome analysis, namely the non-genotropic androgen signaling and the HIF1alpha pathway. We provide evidence that p38 MAPK and PI3K/Akt/NFkappaB and/or Rho/Actin pathways are directly involved in testosterone-induced apoptosis, while the JNK/c-JUN pathway is involved in membrane site-initiated transcription. Furthermore we show that membrane-acting androgens modify the transcription of the erythropoietin receptor (EPOR), leading to erythropoietin-initiated actions. Interestingly, association of recombinant human erythropoietin (rHuEPO) together with testosterone-BSA protects cells from apoptosis, through discrete signaling events. The effect of testosterone-BSA is exerted through the classical erythropoietin promoter, while rHuEPO decreases the transcription of EPOR acting on a newly identified regulatory/promoter region, upstream of its known promoter. These results suggest a new interaction of membrane-acting androgen with EPOR and should be taken into account in the pharmaceutical manipulations of breast cancer patients.
Collapse
Affiliation(s)
- Vassiliki Pelekanou
- Laboratory of Experimental Endocrinology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion GR-71003, Greece
| | - George Notas
- Laboratory of Experimental Endocrinology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion GR-71003, Greece
| | - Elias Sanidas
- Department of Surgical Oncology, University of Crete, School of Medicine, Heraklion GR-71003, Greece
| | - Andreas Tsapis
- INSERM, U976, Paris, F-75010 France
- INSERM, Univ Paris-Diderot, Paris, F-75013 France
| | - Elias Castanas
- Laboratory of Experimental Endocrinology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion GR-71003, Greece
| | - Marilena Kampa
- Laboratory of Experimental Endocrinology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion GR-71003, Greece
| |
Collapse
|
28
|
Geisler J, Suzuki T, Helle H, Miki Y, Nagasaki S, Duong NK, Ekse D, Aas T, Evans DB, Lønning PE, Sasano H. Breast cancer aromatase expression evaluated by the novel antibody 677: correlations to intra-tumor estrogen levels and hormone receptor status. J Steroid Biochem Mol Biol 2010; 118:237-41. [PMID: 19883757 DOI: 10.1016/j.jsbmb.2009.10.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 10/19/2009] [Accepted: 10/21/2009] [Indexed: 11/28/2022]
Abstract
Breast cancer tissue estrogen levels on an average exceed plasma as well as benign breast tissue levels. To evaluate the contribution of intra-tumor aromatization to individual tumor estrogen levels (estradiol, E2; estrone, E1; estrone sulfate, E1S), breast cancer tissue sections obtained during mastectomy in 28 postmenopausal breast cancer patients were stained for aromatase protein expression using the aromatase antibody 677. The findings were correlated to intra-tumor estrogen levels determined with a highly sensitive HPLC-RIA. Staining with 677 alone (irrespective of the hormone receptor status) revealed no difference in tumor E2 levels comparing 677+ versus 677- tumors, although a non-significant trend towards higher tumor E1 and E1S levels was observed in 677+ breast cancers. In contrast, tumor levels of E(2) were significantly higher in ER+ tumors compared to ER- tumors (P<0.001) and to benign breast tissue from the same breast (P<0.001). Analysing the additional effect of positive staining with the aromatase antibody 677 on tumor estrogen levels in the subgroup of ER+ tumors, revealed significantly higher tumor levels of E2 (mean level of 544.7 versus 197.1 fmol/g tissue) as well as a non-significant trend concerning tumor E1 (mean level of 296.9 versus 102.1 fmol/g tissue). The mean tumor tissue E1S level was observed somewhat lower in ER+677+ (103.5 fmol/g) versus ER+677- tumors (190.1 fmol/g). In the subgroup of ER+PgR+ tumors, tissue levels of E2 were also found to be significantly higher among 677+ compared to 677- tumors: 873.2 fmol/g (95% CI 395.9-1925.6) versus 217.9 fmol/g (95% CI 88.8-534.9) (P=0.015). In conclusion, our results indicate a moderate effect of aromatase enzyme expression evaluated by IHC using the antibody 677 on intra-tumor estrogen levels among ER+ breast cancers. A substantial interindividual variation in the ratios between the individual estrogen fractions suggests additional effects, like alterations in other enzymes to be involved in the intra-tumor estrogen homeostasis.
Collapse
Affiliation(s)
- Jürgen Geisler
- Section of Oncology, Institute of Medicine, University of Bergen, N-5021 Bergen, Norway.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Dietel M. Hormone replacement therapy (HRT), breast cancer and tumor pathology. Maturitas 2009; 65:183-9. [PMID: 20005648 DOI: 10.1016/j.maturitas.2009.11.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 10/25/2009] [Accepted: 11/02/2009] [Indexed: 11/28/2022]
Abstract
Within an average observation period of 5-6 years, several clinical trials reported an increased risk of breast cancer due to hormone replacement therapy (HRT). However, it remains disputable, whether the increased rate of breast cancers detected within the given time frame is indeed due to newly induced tumors and thus constitutes HRT-initiated primary breast cancers. Onco-pathologically speaking it appears more likely that HRT stimulates the growth of already existing small tumor nests which - due to their small size - would otherwise go undiagnosed. The major arguments are: In summary, HRT is hence more likely to be a tumor promoter than a de novo-inducer of breast cancers.
Collapse
Affiliation(s)
- Manfred Dietel
- Institut für Pathologie, Charité, Campus Mitte, Humboldt Universität Berlin, Charitéplatz 1, D-10117 Berlin, Germany.
| |
Collapse
|
30
|
Lønning PE, Helle H, Duong NK, Ekse D, Aas T, Geisler J. Tissue estradiol is selectively elevated in receptor positive breast cancers while tumour estrone is reduced independent of receptor status. J Steroid Biochem Mol Biol 2009; 117:31-41. [PMID: 19591931 DOI: 10.1016/j.jsbmb.2009.06.005] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 06/24/2009] [Accepted: 06/26/2009] [Indexed: 01/29/2023]
Abstract
Previous studies have suggested elevated estrogen production in tumour-bearing breast quadrants as well as in breast cancers versus benign tissue. Using highly sensitive assays, we determined breast cancer tissue estrogen concentrations together with plasma and benign tissue estrogen concentrations in each quadrant obtained from mastectomy specimens (34 postmenopausal and 13 premenopausal women). We detected similar concentrations of each of the three major estrogens estradiol (E(2)), estrone (E(1)) and E(1)S in tumour-bearing versus non-tumour-bearing quadrants. Considering malignant tumours, intratumour E(1) levels were reduced in cancer tissue obtained from pre- as well as postmenopausal women independent of tumour ER status (average ratio E(1) cancer: benign tissue of 0.2 and 0.3, respectively; p<0.001 for both groups), suggesting intratumour aromatization to be of minor importance. The most striking finding was a significant (4.1-8.6-fold) increased E(2) concentration in ER positive tumours versus normal tissue (p<0.05 and <0.001 for pre- and postmenopausal patients, respectively), contrasting low E(2) concentrations in ER- tumours (p<0.01 and <0.001 comparing E(2) levels between ER+ and ER- tumours in pre- and postmenopausals, respectively). A possible explanation to our finding is increased ligand receptor binding capacity for E(2) in receptor positive tumours but alternative factors influencing intratumour estrogen disposition cannot be excluded.
Collapse
Affiliation(s)
- P E Lønning
- Section of Oncology, Institute of Medicine, University of Bergen, Jonas Lies vei 26, N-5021 Bergen, Norway.
| | | | | | | | | | | |
Collapse
|
31
|
Neilson HK, Friedenreich CM, Brockton NT, Millikan RC. Physical activity and postmenopausal breast cancer: proposed biologic mechanisms and areas for future research. Cancer Epidemiol Biomarkers Prev 2009; 18:11-27. [PMID: 19124476 DOI: 10.1158/1055-9965.epi-08-0756] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Convincing evidence now supports a probable preventive role for physical activity in postmenopausal breast cancer. The mechanisms by which long-term physical activity affect risk, however, remain unclear. The aims of this review were to propose a biological model whereby long-term physical activity lowers postmenopausal breast cancer risk and to highlight gaps in the epidemiologic literature. To address the second aim, we summarized epidemiologic literature on 10 proposed biomarkers, namely, body mass index (BMI), estrogens, androgens, sex hormone binding globulin, leptin, adiponectin, markers of insulin resistance, tumor necrosis factor-alpha, interleukin-6, and C-reactive protein, in relation to postmenopausal breast cancer risk and physical activity, respectively. Associations were deemed "convincing," "probable," "possible," or "hypothesized" using set criteria. Our proposed biological model illustrated the co-occurrence of overweight/obesity, insulin resistance, and chronic inflammation influencing cancer risk through interrelated mechanisms. The most convincing epidemiologic evidence supported associations between postmenopausal breast cancer risk and BMI, estrogens, and androgens, respectively. In relation to physical activity, associations were most convincing for BMI, estrone, insulin resistance, and C-reactive protein. Only BMI and estrone were convincingly (or probably) associated with both postmenopausal breast cancer risk and physical activity. There is a need for prospective cohort studies relating the proposed biomarkers to cancer risk and for long-term exercise randomized controlled trials comparing biomarker changes over time, specifically in postmenopausal women. Future etiologic studies should consider interactions among biomarkers, whereas exercise trials should explore exercise effects independently of weight loss, different exercise prescriptions, and effects on central adiposity.
Collapse
Affiliation(s)
- Heather K Neilson
- Division of Population Health, Alberta Cancer Board, 1331-29 Street NW, Calgary, Alberta, Canada T2N 4N2
| | | | | | | |
Collapse
|
32
|
Gogoi R, Kudla M, Gil O, Fishman D. The activity of medroxyprogesterone acetate, an androgenic ligand, in ovarian cancer cell invasion. Reprod Sci 2009; 15:846-52. [PMID: 19017820 DOI: 10.1177/1933719108323446] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVES An epithelial ovarian cancer cell line constitutively expressing the androgen receptor was created to evaluate the mechanism and effects of androgen receptor activation on epithelial ovarian cancer cell invasion. METHODS Immunocytochemistry and Western blot analyses confirmed androgen receptor expression. Boyden chamber invasion assays were performed using cells treated with the androgen receptor ligands medroxyprogesterone acetate or dihydrotestosterone. The matrix metalloproteinases associated with invasion were investigated using zymographic assays. RESULTS Androgen receptor-mediated invasion is ligand dependent. While both medroxyprogesterone acetate and dihydrotestosterone signal through androgen receptor, medroxyprogesterone acetate is more effective at stimulating invasion of epithelial ovarian cancer cells. Unlike the wild-type epithelial ovarian cancer cells, this increase in invasion in androgen receptor + epithelial ovarian cancer cells does not seem to be dependent on matrix metalloproteinase 2 or 9 activation. CONCLUSION Although classified as a progestin, medroxyprogesterone acetate has significant androgenic activity unique from the pure androgen dihydrotestosterone. Our studies suggest that pharmacologic doses of medroxyprogesterone acetate may actually increase the invasive potential of epithelial ovarian cancer cells.
Collapse
Affiliation(s)
- Radhika Gogoi
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, New York University, New York, NY, USA
| | | | | | | |
Collapse
|
33
|
Tiefenbacher K, Daxenbichler G. The Role of Androgens in Normal and Malignant Breast Tissue. Breast Care (Basel) 2008; 3:325-331. [PMID: 20824027 DOI: 10.1159/000158055] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Androgens, like estrogens, can be synthesized in the breast. As both active androgens and their corresponding receptors are present in breast tissue, we conclude that they play a role in breast physiology. This is supported by the fact that insufficient androgen production or sensitivity results in the development of gynecomastia. Complete androgen insensitivity due to receptor defects leads to normal female breast development in these XY women. While breast development is completely inhibited by male testosterone levels, partial but not total degradation of a developed breast by androgen treatment appears to be possible. Breast cancer in early stages seems to fulfill the prerequisites of androgen responsiveness. Androgen treatment of advanced breast cancer has shown similar effectiveness as anti-estrogen or estrogen-ablative therapy, but also considerable side effects. It has been speculated that the use of selective androgen modulators (SARMs), either alone or preferably in addition to anti-estrogens or aromatase inhibitors, may be a promising alternative to current therapy modalities in hormone-dependent breast cancer. In addition, future studies on the use of SARMs in prophylactic settings seem to be justified.
Collapse
|
34
|
Intratumoral estrogen production in breast carcinoma: significance of aromatase. Breast Cancer 2008; 15:270-7. [DOI: 10.1007/s12282-008-0062-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Accepted: 05/12/2008] [Indexed: 10/21/2022]
|
35
|
Suzuki T, Miki Y, Akahira JI, Moriya T, Ohuchi N, Sasano H. Aromatase in human breast carcinoma as a key regulator of intratumoral sex steroid concentrations. Endocr J 2008; 55:455-63. [PMID: 18480557 DOI: 10.1507/endocrj.k07e-053] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
It is well-known that estrogens are closely involved in the growth of human breast carcinomas, and that the great majority of breast carcinoma express estrogen receptors. Recent studies have demonstrated that estrogens are locally produced and act on the breast carcinoma tissue. Among these pathways, aromatase is a key enzyme for intratumoral production of estrogens in breast carcinomas, and aromatase inhibitors are currently used in the breast carcinoma in postmenopausal women as an estrogen deprivation therapy. This review summarizes the results of recent studies on the expression and regulation of aromatase in breast carcinoma tissues, and discusses the potential biological and/or clinical significance of aromatase. Aromatase is abundantly expressed in various cell types, such as carcinoma cells, intratumoral stromal cells, and adipocytes adjacent to the carcinoma, in breast carcinoma tissues. Further, a key regulator for aromatase expression differed according to cell type. In addition, aromatase suppressed in situ production of bioactive androgen, 5alpha-dihydrotestosterone (DHT), in breast carcinoma. Aromatase inhibitors may thus have additional antiproliferative effects through increasing local DHT concentration with estrogen deprivation.
Collapse
Affiliation(s)
- Takashi Suzuki
- Department of Pathology, Tohoku University School of Health Sciences, Sendai, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Intratumoral metabolism and synthesis of biologically active steroids such as estradiol and 5alpha-dihydrotestosterone as a result of interactions of various enzymes are considered to play very important roles in the pathogenesis and development of hormone-dependent breast carcinoma. Among these enzymes involved in estrogen metabolism, intratumoral aromatase play an important role in converting androgens to estrogens in situ from serum and serving as the source of estrogens, especially in postmenopausal patients with breast carcinoma. However, other enzymes such as 17beta-hydroxysteroid dehydrogenase (17beta-HSD) isozymes, estrogen sulfatase (STS), and estrogen sulfotransferase, which contribute to in situ availability of biologically active estrogens, also play pivotal roles in this intratumoral estrogen production above. Androgen action on human breast carcinoma has not been well-studied but are considered important not only in hormonal regulation but also other biological features of carcinoma cells. Intracrine mechanisms also play important roles in androgen actions on human breast carcinoma cells. Among the enzymes involved in biologically active androgen metabolism and/or synthesis, both 17beta-hydroxysteroid dehydrogenase type 5 (17betaHSD5; conversion from circulating androstenedione to testosterone) and 5alpha-reductase (5alphaRed; reduction of testosterone to DHT (5alpha-dihydrotestosterone) were expressed in breast carcinoma tissues, and in situ production of DHT has been proposed in human breast cancer tissues. However, intracrine mechanisms of androgens as well as their biological or clinical significance in the patients with breast cancer have not been fully elucidated in contrast to those in estrogens.
Collapse
Affiliation(s)
- Hironobu Sasano
- Department of Pathology, Tohoku University School of Medicine, 2-1 Seiryou-machi, Aoba-ku, Sendai, Japan.
| | | | | | | |
Collapse
|
37
|
Endogenous Hormone Levels and Risk of Breast, Endometrial and Ovarian Cancers:. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008. [DOI: 10.1007/978-0-387-78818-0_10] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
38
|
Development of a high sensitivity, nested Q-PCR assay for mouse and human aromatase. Breast Cancer Res Treat 2007; 111:343-51. [PMID: 17975728 DOI: 10.1007/s10549-007-9792-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Accepted: 10/09/2007] [Indexed: 10/22/2022]
Abstract
Measurement of breast tissue estradiol levels could provide a powerful method to predict the risk of developing breast cancer but obtaining sufficient amounts of tissue from women is difficult from a practical standpoint. Assessment of aromatase in ductal lavage fluid or fine needle aspirates from breast might provide a surrogate marker for tissue estrogen levels but highly sensitive methods would be required. These considerations prompted us to develop an ultra-sensitive, "nested" PCR assay for aromatase which is up to one million fold more sensitive than standard PCR methods. We initially validated this assay using multiple tissues from the aromatase transgenic mouse and found that coefficients of variation for measurement of replicate samples averaged less than 5%. We demonstrated a 60-fold enhancement in aromatase message in the transgenic versus the wild type mouse breast but surprisingly, levels in the transgenic animals were highly variable, ranging from 0.4 to 27 relative units. The variability of aromatase expression in the transgenic breast did not correlate with the degree of breast development and did not appear to relate to hormonal manipulation of the MMTV promoter but probably related to lack of exhaustive inbreeding and mixed zygocity of transgenic animals. Extensive validation in mouse tissues provided confidence regarding the assay in human tissues, since nearly identical methods were used. The human assay was sufficiently sensitive to detect aromatase in a single human JAR (choriocarcinoma) cell, in all breast biopsies measured, and in 7/23 ductal lavage fluids.
Collapse
|
39
|
Abstract
Progestins exert their progestational activity by binding to the progesterone receptor (form A, the most active and form B, the less active) and may also interact with other steroid receptors (androgen, glucocorticoid, mineralocorticoid, estrogen). They can have important effects in other tissues besides the endometrium, including the breast, liver, bone and brain. The biological responses of progestins cover a very large domain: lipids, carbohydrates, proteins, water and electrolyte regulation, hemostasis, fibrinolysis, and cardiovascular and immunological systems. At present, more than 200 progestin compounds have been synthesized, but the biological response could be different from one to another depending on their structure, metabolism, receptor affinity, experimental conditions, target tissue or cell line, as well as the biological response considered. There is substantial evidence that mammary cancer tissue contains all the enzymes responsible for the local biosynthesis of estradiol (E(2)) from circulating precursors. Two principal pathways are implicated in the final steps of E(2) formation in breast cancer tissue: the 'aromatase pathway', which transforms androgens into estrogens, and the 'sulfatase pathway', which converts estrone sulfate (E(1)S) into estrone (E(1)) via estrone sulfatase. The final step is the conversion of weak E(1) to the potent biologically active E(2) via reductive 17beta-hydroxysteroid dehydrogenase type 1 activity. It is also well established that steroid sulfotransferases, which convert estrogens into their sulfates, are present in breast cancer tissues. It has been demonstrated that various progestins (e.g. nomegestrol acetate, medrogestone, promegestone) as well as tibolone and their metabolites can block the enzymes involved in E(2) bioformation (sulfatase, 17beta-hydroxysteroid dehydrogenase) in breast cancer cells. These substances can also stimulate the sulfotransferase activity which converts estrogens into the biologically inactive sulfates. The action of progestins in breast cancer is very controversial; some studies indicate an increase in breast cancer incidence, others show no difference and still others a significant decrease. Progestin action can also be a function of combination with other molecules (e.g. estrogens). In order to clarify and better understand the response of progestins in breast cancer (incidence, mortality), as well as in hormone replacement therapy or endocrine dysfunction, new clinical trials are needed studying other progestins as a function of the dose and period of treatment.
Collapse
Affiliation(s)
- Jorge R Pasqualini
- Hormones and Cancer Research Unit, Institut de Puériculture et de Périnatalogie, Paris, France.
| |
Collapse
|
40
|
Suzuki T, Miki Y, Moriya T, Akahira JI, Hirakawa H, Ohuchi N, Sasano H. In situ production of sex steroids in human breast carcinoma. Med Mol Morphol 2007; 40:121-7. [PMID: 17874044 DOI: 10.1007/s00795-007-0365-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Accepted: 03/06/2007] [Indexed: 10/22/2022]
Abstract
It is well known that sex steroids are closely involved in the growth of human breast carcinomas, and the great majority of breast carcinomas express sex steroid receptors. In particular, recent studies have demonstrated that estrogens and androgens are locally produced and act in breast carcinoma tissues without release into plasma. Blockade of intratumoral estrogen production potentially leads to an improvement in the prognosis of invasive breast carcinoma patients, and, therefore, it is important to obtain a better understanding of sex steroid-producing enzymes in breast carcinoma. In this review, we summarize recent studies on tissue concentration of sex steroids and expression of enzymes related to intratumoral production of estrogens [aromatase, steroid sulfatase (STS), and 17beta-hydroxysteroid dehydrogenase type 1 (17betaHSD1)], and androgens (17betaHSD5 and 5alpha-reductase) in invasive and in situ (noninvasive) breast carcinomas, and discuss the significance of intratumoral production of sex steroids in breast carcinoma.
Collapse
Affiliation(s)
- Takashi Suzuki
- Department of Pathology, Tohoku University School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | | | | | | | | | | | | |
Collapse
|
41
|
Hankinson SE, Eliassen AH. Endogenous estrogen, testosterone and progesterone levels in relation to breast cancer risk. J Steroid Biochem Mol Biol 2007; 106:24-30. [PMID: 17719770 PMCID: PMC2715949 DOI: 10.1016/j.jsbmb.2007.05.012] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Multiple lines of evidence support a central role of hormones in the etiology of breast cancer. In epidemiologic studies, considerable effort has focused on delineating the role of endogenous hormones in risk of breast cancer among postmenopausal women. Recently, substantial additional data has accrued from prospective studies where endogenous hormones are measured in study subjects prior to disease diagnosis. In this review, the epidemiologic evidence linking sex steroids--estrogens, testosterone, and progesterone, specifically--with subsequent risk of breast cancer in both premenopausal and postmenopausal women is summarized. Overall, a strong positive association between breast cancer risk and circulating levels of both estrogens and testosterone has now been well confirmed among postmenopausal women; women with hormone levels in the top 20% of the distribution (versus bottom 20%) have a two- to three-fold higher risk of breast cancer. Evidence among premenopausal women is more limited, though increased risk associated with higher levels of testosterone is consistent. However, both positive and null associations have been observed with estrogens and progesterone and clearly more evaluation is needed.
Collapse
Affiliation(s)
- Susan E Hankinson
- Channing Laboratory, Department of Medicine, 181 Longwood Avenue, Harvard Medical School, Brigham and Women's Hospital, MA 02115, USA.
| | | |
Collapse
|
42
|
Tamimi RM, Byrne C, Colditz GA, Hankinson SE. Endogenous hormone levels, mammographic density, and subsequent risk of breast cancer in postmenopausal women. J Natl Cancer Inst 2007; 99:1178-87. [PMID: 17652278 DOI: 10.1093/jnci/djm062] [Citation(s) in RCA: 181] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Mammographic density and circulating sex hormones are two well-confirmed predictors of breast cancer risk. Whether mammographic density reflects levels of endogenous sex hormones is unclear. We examined whether these predictors are independently associated with breast cancer risk in a prospective study. METHODS We conducted a nested case-control study within the Nurses' Health Study cohort of 253 case subjects with breast cancer and 520 control subjects. All participants were postmenopausal women who were not using postmenopausal hormones at the time of both blood collection and mammography. Plasma levels of estradiol, free estradiol, testosterone, and free testosterone were evaluated. Mammographic density was assessed by use of computer-assisted analysis of mammograms. Logistic regression models that were adjusted for matching variables and potential confounders were used to calculate relative risks (RRs) and 95% confidence intervals (CIs). All statistical tests were two-sided. RESULTS Levels of circulating sex steroids and mammographic density were both statistically significantly and independently associated with breast cancer risk. The relative risk of breast cancer associated with mammographic density (RR for highest quartile compared with lowest quartile = 3.8, 95% CI = 2.2 to 6.6; P(trend)<.001) changed little when the analysis was adjusted for circulating estradiol (RR = 3.9, 95% CI = 2.2 to 6.9; P(trend)<.001) or circulating testosterone (RR = 4.1, 95% CI = 2.3 to 7.2; P(trend)<.001). Circulating levels of estradiol (RR = 2.4, 95% CI = 1.4 to 4.0) and of testosterone (RR = 2.0, 95% CI = 1.2 to 3.1) were both associated with breast cancer risk, before and after adjustment for mammographic density. In a joint analysis of mammographic density and plasma testosterone, the risk of breast cancer was highest in the highest tertiles of both relative to the lowest tertiles of both (RR = 6.0, 95% CI = 2.6 to 14.0). A similar pattern was observed in the joint analysis of estradiol and mammographic density (RR = 4.1, 95% CI = 1.7 to 9.8). CONCLUSIONS Circulating sex steroid levels and mammographic density appear strongly and independently associated with the risk of breast cancer in postmenopausal women.
Collapse
Affiliation(s)
- Rulla M Tamimi
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Ave, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
43
|
Nicolás Díaz-Chico B, Germán Rodríguez F, González A, Ramírez R, Bilbao C, Cabrera de León A, Aguirre Jaime A, Chirino R, Navarro D, Díaz-Chico JC. Androgens and androgen receptors in breast cancer. J Steroid Biochem Mol Biol 2007; 105:1-15. [PMID: 17631997 DOI: 10.1016/j.jsbmb.2006.11.019] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2006] [Accepted: 11/09/2006] [Indexed: 11/28/2022]
Abstract
Aromatase (CYP19) converts adrenal and ovarian androgens into estrogens, which supports the growth of estrogen-dependent breast cancers. Anti-aromatase agents are displacing antiestrogens as the first-line treatment for estrogen receptor positive breast cancers. Androgens can act as estrogen precursors, but besides this capability they can also directly act on breast cancer cells by binding to androgen receptors, which are present in the majority of breast cancer specimens. Epidemiological and clinical evidences suggest that higher levels of circulating androgen increase the risk of developing breast cancer. Androgen receptor gene polymorphisms which render the more transcriptionally active receptors have been related to a lower risk of breast cancer. It is currently accepted that androgens act as antiproliferative agents in the presence of estrogens in some breast cancer cell lines. However, emerging evidence suggests that direct androgenic activity might also stimulate cell growth in a subset of estrogen-resistant breast tumors. Here we discuss the supporting evidence which proposes that androgens themselves are actively involved in breast carcinogenesis and its clinical behaviour.
Collapse
|
44
|
Macedo LF, Guo Z, Tilghman SL, Sabnis GJ, Qiu Y, Brodie A. Role of androgens on MCF-7 breast cancer cell growth and on the inhibitory effect of letrozole. Cancer Res 2006; 66:7775-82. [PMID: 16885381 DOI: 10.1158/0008-5472.can-05-3984] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Previous work has shown that androgens inhibit breast cancer cells and tumor growth. On the other hand, androgens can be converted to mitogenic estrogens by aromatase in breast cancer cells. Here, we report that androgens, such as the aromatizable androstenedione and the non-aromatizable 5alpha-dihydrotestosterone, inhibit MCF-7 cell proliferation. This effect is observed only in the absence or at a low concentration of estrogens and is evident in cells with low aromatase activity. Growth of a new aromatase stably transfected MCF-7 cell line (Ac1) was stimulated by conversion of androstenedione into estrogens and was sensitive to aromatase inhibitors. We show that blockade of the androgen receptor (AR) in these cells by the antiandrogen casodex or by the anti-AR small interfering RNA inhibited the antiproliferative effect of dihydrotestosterone and letrozole (aromatase inhibitor). We also show that suppression of the estrogen-induced antiapoptotic protein Bcl-2 may be involved in the antiproliferative effects of androgens and letrozole. These effects can be reversed by casodex. In conclusion, the results suggest that aromatase inhibitors may exert their antiproliferative effect not only by reducing the intracellular production of estrogens but also by unmasking the inhibitory effect of androgens acting via the AR.
Collapse
Affiliation(s)
- Luciana F Macedo
- Department of Pharmacology and Experimental Therapeutics, University of Maryland, 685 West Baltimore Street, Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
45
|
Beattie MS, Costantino JP, Cummings SR, Wickerham DL, Vogel VG, Dowsett M, Folkerd EJ, Willett WC, Wolmark N, Hankinson SE. Endogenous sex hormones, breast cancer risk, and tamoxifen response: an ancillary study in the NSABP Breast Cancer Prevention Trial (P-1). J Natl Cancer Inst 2006; 98:110-5. [PMID: 16418513 DOI: 10.1093/jnci/djj011] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Prospective studies have shown an association between increased serum levels of estradiol and testosterone and breast cancer risk in postmenopausal women. Raloxifene has been shown to reduce breast cancer risk more in women with high estradiol levels than in those with lower levels. The purpose of this study was to determine whether sex hormone levels were associated with breast cancer risk and with response to tamoxifen in a high-risk population. METHODS Using a case-cohort design, we studied 135 women with postmenopausal breast cancer and 275 postmenopausal women without breast cancer who were enrolled in the National Surgical Adjuvant Breast and Bowel Project Cancer Prevention Trial (P-1) and who had been treated with tamoxifen or placebo for 69 months. We measured estradiol, testosterone, and sex hormone-binding globulin by using radioimmunoassay in baseline plasma samples. Relative risks (RRs) and 95% confidence intervals (CIs) for invasive breast cancer were estimated for each quartile of sex hormone level using Cox proportional hazards models. All statistical tests were two-sided. RESULTS Median plasma levels of estradiol, testosterone, and sex hormone-binding globulin were similar between the case and cohort groups. The relative risk of breast cancer for women in the placebo group was not associated with sex hormone levels (risk of estrogen receptor-positive breast cancer in women by quartile of estradiol: Q1 [lowest], RR = 1.0; Q2, RR = 1.16, 95% CI = 0.49 to 2.7; Q3, RR = 1.08, 95% CI = 0.45 to 2.61; and Q4, RR = 1.29, 95% CI = 0.59 to 2.82). The reduced risk of invasive breast cancer in tamoxifen-treated women compared with placebo-treated women was not associated with sex hormone levels. CONCLUSIONS These data do not support the use of endogenous sex hormone levels to identify women who are at particularly high risk of breast cancer and who are most likely to benefit from chemoprevention with tamoxifen.
Collapse
Affiliation(s)
- Mary S Beattie
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, CA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Tamimi RM, Hankinson SE, Colditz GA, Byrne C. Endogenous sex hormone levels and mammographic density among postmenopausal women. Cancer Epidemiol Biomarkers Prev 2006; 14:2641-7. [PMID: 16284390 DOI: 10.1158/1055-9965.epi-05-0558] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Mammographic density is one of the strongest predictors of breast cancer risk. The mechanism by which breast density increases breast cancer risk is unclear although it has been hypothesized that breast density reflects cumulative exposure to estrogens. METHODS To evaluate this hypothesis, we conducted a cross-sectional study among 520 postmenopausal women in the Nurses' Health Study that examined the relation between circulating sex hormones and mammographic density. Women were postmenopausal and not taking exogenous hormones at the time of blood collection and mammogram. Percent breast density was measured from digitized mammograms using a computer-assisted method. Circulating estrone, estradiol, androstenedione, testosterone, DHEA, DHEA sulfate, sex hormone-binding globulin, progesterone, and prolactin were measured in plasma. RESULTS In contrast to the prior hypothesis, circulating estrogens were inversely related to percent mammographic density. The mean percent mammographic density was 25.6% among women in the lowest quartile of circulating estradiol compared with 14.4% among women in the highest quartile [Spearman correlation (r) = -0.22, P < 0.0001]. Circulating estrogens alone explained 1% to 5% of the variation of mammographic density. Body mass index was positively associated with circulating estradiol levels (r = 0.45, P < 0.0001) and inversely related to percent mammographic density (r = -0.51, P < 0.0001). After adjustment for body mass index, there was no association between estradiol and breast density (r = 0.01, P = 0.81). Likewise, there was no relation between the other sex hormones measured or prolactin and mammographic density after adjustment for body mass index. CONCLUSION These findings indicate that in postmenopausal women, mammographic density is independent of circulating sex hormone levels.
Collapse
Affiliation(s)
- Rulla M Tamimi
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
47
|
Suzuki T, Miki Y, Moriya T, Akahira JI, Ishida T, Hirakawa H, Yamaguchi Y, Hayashi SI, Sasano H. 5α-Reductase type 1 and aromatase in breast carcinoma as regulators ofin situ androgen production. Int J Cancer 2006; 120:285-91. [PMID: 17066438 DOI: 10.1002/ijc.22317] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Previous in vitro studies demonstrated that bioactive androgen 5alpha-dihydrotestosterone (DHT) exerted antiproliferative effects through an interaction with androgen receptor (AR) in breast carcinoma cells. However, AR status has not been examined in association with DHT concentration in breast carcinoma tissues, and significance of androgenic actions remains unclear in breast carcinomas. Therefore, in our study, we first examined intratumoral DHT concentrations in 38 breast carcinoma tissues using liquid chromatography/electrospray tandem mass spectrometry. Intratumoral DHT concentration was positively associated with 5alpha-reductase type 1 (5alphaRed1), and negatively correlated with aromatase. We then examined clinical significance of AR and 5alphaRed1 status in 115 breast carcinoma tissues by immunohistochemistry. Breast carcinomas positive for both AR and 5alphaRed1 were inversely associated with tumor size or Ki-67. These patients showed significant associations with a decreased risk of recurrence and improved prognosis for overall survival, and the AR / 5alphaRed1 status was demonstrated an independent prognostic factor. Moreover, we examined possible regulation of DHT production by aromatase in in vitro studies. DHT synthesis from androstenedione in MCF-7 cells was significantly inhibited by coculture with aromatase-positive stromal cells, which was significantly reversed by addition of aromatase inhibitor exemestane. These results suggest that intratumoral DHT concentration is mainly determined by 5alphaRed1 and aromatase in breast carcinoma tissues, and antiproliferative effect of DHT may primarily occur in the cases positive for both AR and 5alphaRed1. Aromatase inhibitors may be more effective in these patients, possibly due to increasing local DHT concentration with estrogen deprivation.
Collapse
MESH Headings
- 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/analysis
- 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism
- Adult
- Aged
- Aged, 80 and over
- Androgens/analysis
- Androgens/metabolism
- Aromatase/analysis
- Aromatase/drug effects
- Aromatase/metabolism
- Aromatase Inhibitors/pharmacology
- Breast Neoplasms/chemistry
- Breast Neoplasms/enzymology
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/chemistry
- Carcinoma, Ductal, Breast/enzymology
- Carcinoma, Ductal, Breast/pathology
- Chromatography, Liquid
- Coculture Techniques
- Dihydrotestosterone/analysis
- Dihydrotestosterone/metabolism
- Female
- Humans
- Immunohistochemistry
- Ki-67 Antigen/analysis
- Middle Aged
- Receptors, Androgen/analysis
- Receptors, Androgen/metabolism
- Tandem Mass Spectrometry
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Takashi Suzuki
- Department of Pathology, Tohoku University School of Medicine, Sendai, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Cummings SR, Lee JS, Lui LY, Stone K, Ljung BM, Cauleys JA. Sex hormones, risk factors, and risk of estrogen receptor-positive breast cancer in older women: a long-term prospective study. Cancer Epidemiol Biomarkers Prev 2005; 14:1047-51. [PMID: 15894651 DOI: 10.1158/1055-9965.epi-04-0375] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Antiestrogens reduce the risk of estrogen receptor-positive (ER+) but not ER-negative (ER-) breast cancer. Women at high risk of ER+ cancer would be the most likely to benefit from these treatments, but the best approach to predicting ER+ cancer is uncertain. METHODS We prospectively assessed putative risk factors for breast cancer and archived serum at -190 degrees C from a community-based cohort of 7,676 women ages > or =65 years who had no history of breast cancer. Follow-up for breast cancer over 10.5 years was 99% complete. Using a case-cohort design, we measured baseline levels of estradiol and testosterone in 196 cases of invasive ER+ cancer and 378 randomly selected controls. RESULTS Women whose testosterone level in highest two quintiles had a 4-fold increased risk of ER+ breast cancer (P < 0.0001). High estradiol concentration also indicated an increased risk but was not a significant predictor after adjustment for testosterone. Women with >16 years of education had a 2.1 times increased risk (P = 0.03) of ER+ cancer, but no other risk factors were significantly related to an increased risk of ER+ cancer. Women with a family history of breast cancer had a 2.9-fold increased risk of ER- cancer (P = 0.002) but no increased risk of ER+ cancer (relative hazard = 1.2, 0.8-1.8). CONCLUSIONS High serum testosterone and advanced education predicted ER+ breast cancer. If confirmed, high testosterone level may be more accurate than family history of breast cancer and other conventional risk factors for identifying older women who are most likely to benefit from antiestrogen chemoprevention.
Collapse
Affiliation(s)
- Steven R Cummings
- California Pacific Medical Center, Coordinating Center, Suite 600, 74 New Montgomery Street, San Francisco, CA 94105, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Rubolini D, Romano M, Martinelli R, Saino N. Effects of elevated yolk testosterone levels on survival, growth and immunity of male and female yellow-legged gull chicks. Behav Ecol Sociobiol 2005. [DOI: 10.1007/s00265-005-0057-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
50
|
Abstract
In recent years, increased attention to women's sexual health has propelled basic scientific research and clinical trials investigating treatment paradigms for improving sexual well-being. As the prevalence of female sexual dysfunction has become manifest, knowledge of the intricate pathophysiological role of androgens in maintaining sexual function has fostered a clearer understanding of the effect of age on androgen status, the role of androgens in the postmenopausal ovary, and aetiological mechanisms of androgen insufficiency in premenopausal and postmenopausal women. Understanding the long-term safety and efficacy of physiological androgen replacement and the development of sensitive testosterone assays for specific use in women will better characterise women who are most likely to respond to androgen therapy and, thereby, optimise their quality of life.
Collapse
Affiliation(s)
- Crista E Johnson
- Female Sexual Medicine Center, Department of Urology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90024, USA.
| | | |
Collapse
|