1
|
Gurrala RR, Kumar T, Yoo A, Mundinger GS, Womac DJ, Lau FH. The Impact of Exogenous Testosterone on Breast Cancer Risk in Transmasculine Individuals. Ann Plast Surg 2023; 90:96-105. [PMID: 36534108 DOI: 10.1097/sap.0000000000003321] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Exogenous testosterone is vital to gender-affirming therapy for transmasculine individuals. Testosterone may be implicated in breast cancer (BCa) because it can activate androgen and estrogen receptors. To further explore this risk, we performed a systematic review to investigate the impact of exogenous testosterone on BCa risk in transmasculine individuals. METHODS We searched PubMed/MEDLINE and Ovid/Embase for clinical and preclinical studies assessing BCa and testosterone therapy and screened 6125 articles independently. We ascertained level of evidence using a modified tool from Cook et al (Chest. 1992;102:305S-311S) and risk of bias using a modified Joanna Briggs Institute's Critical Appraisal Tool. RESULTS Seventy-six studies were included. Epidemiological data suggested that BCa incidence was higher in transmasculine individuals compared with cisgender men but lower compared with cisgender women. Histological studies of transmasculine breast tissue samples also demonstrated a low incidence of precancerous lesions. Interestingly, cases demonstrated that BCa occurred at a younger average age in transmasculine individuals and was predominantly hormone receptor positive. The mechanism for BCa in transmasculine individuals may be related to androgen receptor stimulation or conversion to estradiol. Serum studies reported varied estradiol levels associated with exogenous testosterone. Animal and in vitro studies demonstrated that testosterone was growth inhibitory but may induce proliferation at higher doses or with low estradiol levels. CONCLUSIONS Plastic surgeons play a critical role in providing gender-affirming care for transmasculine patients. The limited studies available suggest that this patient population has decreased risk for BCa when compared with cisgender women; however, any BCa that does occur may have different clinical presentations and underlying mechanisms compared with cisgender women and men. Overall, the limitations for clinical studies and discrepancies among preclinical studies warrant further investigation.
Collapse
Affiliation(s)
| | | | - Aran Yoo
- Section of Plastic and Reconstructive Surgery, Department of Surgery, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA
| | | | - Daniel J Womac
- Section of Plastic and Reconstructive Surgery, Department of Surgery, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA
| | - Frank H Lau
- Section of Plastic and Reconstructive Surgery, Department of Surgery, Louisiana State University Health Sciences Center New Orleans, New Orleans, LA
| |
Collapse
|
2
|
Alsawalha L, Ahram M, Abdullah MS, Dalmizrak O. Enzalutamide Overcomes Dihydrotestosterone Induced Chemo-Resistance In Triple-Negative Breast Cancer Cells via Apoptosis. Anticancer Agents Med Chem 2022; 22:3038-3048. [DOI: 10.2174/1871520622666220509123505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/15/2022] [Accepted: 03/15/2022] [Indexed: 11/22/2022]
Abstract
Background:
Triple-negative breast cancer is challenging to treat due to its heterogeneity and lack of therapeutic targets. Hence, systemic chemotherapy is still the mainstay in TNBC treatment. Unfortunately, patients commonly develop chemo-resistance. Androgen signalling through its receptor is an essential player in breast cancer where it has been shown to confer chemo-resistance to TNBC cells
Objective:
To elucidate the mechanistic effects of enzalutamide in the chemoresponse of TNBC cells to doxorubicin through the apoptosis pathway.
Results:
Enzalutamide decreased the viability of MDA-MB-231 and MDA-MB- 453 cells and reduced DHT-induced chemo-resistance of both cell lines. It also increased the chemo-sensitivity towards doxorubicin in MDA-MB-231 cells. Increasing DNA degradation and caspase 3/7 activity were concomitant with these outcomes. Moreover, enzalutamide downregulated the expression of the anti-apoptosis genes, mcl1 and bcl2, in MDA-MB-231 cells. Moreover, increase the pro-apoptotic gene bid. On the other hand, DHT upregulated the expression of the anti-apoptosis genes, mcl1 and bcl2, in both cell lines.
Conclusion:
DHT increases the expression of the anti-apoptosis mcl1 and bcl2 in the TNBC cells, presumably leading to cell survival via the prevention of doxorubicin-induced apoptosis. On the other hand, enzalutamide may sensitize the cells to doxorubicin through downregulation of the bid/bcl2/mcl1 axis that normally activates the executive caspases, caspase 3/7. The activities of the latter enzymes were apparent in DNA degradation at the late stages of
Collapse
Affiliation(s)
| | - Mamoun Ahram
- School of Medicine, The University of Jordan, Amman
| | | | | |
Collapse
|
3
|
Takagi K, Yamaguchi M, Miyashita M, Sasano H, Suzuki T. Diverse role of androgen action in human breast cancer. ENDOCRINE ONCOLOGY (BRISTOL, ENGLAND) 2022; 2:R102-R111. [PMID: 37435447 PMCID: PMC10259322 DOI: 10.1530/eo-22-0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 08/22/2022] [Indexed: 07/13/2023]
Abstract
Breast cancer is a hormone-dependent cancer, and sex steroids play a pivotal role in breast cancer progression. Estrogens are strongly associated with breast cancers, and the estrogen receptor (estrogen receptor α; ERα) is expressed in 70-80% of human breast carcinoma tissues. Although antiestrogen therapies (endocrine therapies) have significantly improved clinical outcomes in ERα-positive breast cancer patients, some patients experience recurrence after treatment. In addition, patients with breast carcinoma lacking ERα expression do not benefit from endocrine therapy. The androgen receptor (AR) is also expressed in >70% of breast carcinoma tissues. Growing evidence supports this novel therapeutic target for the treatment of triple-negative breast cancers that lack ERα, progesterone receptor, and human EGF receptor 2, and ERα-positive breast cancers, which are resistant to conventional endocrine therapy. However, the clinical significance of AR expression is still controversial and the biological function of androgens in breast cancers is unclear. In this review, we focus on the recent findings concerning androgen action in breast cancers and the contributions of androgens to improved breast cancer therapy.
Collapse
Affiliation(s)
- Kiyoshi Takagi
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mio Yamaguchi
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Minoru Miyashita
- Department of Breast and Endocrine Surgical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Pathology, Tohoku University Hospital, Sendai, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
4
|
Jahan N, Jones C, Rahman RL. Androgen receptor expression in breast cancer: Implications on prognosis and treatment, a brief review. Mol Cell Endocrinol 2021; 531:111324. [PMID: 34000352 DOI: 10.1016/j.mce.2021.111324] [Citation(s) in RCA: 6] [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: 11/23/2020] [Revised: 04/28/2021] [Accepted: 05/10/2021] [Indexed: 01/08/2023]
Abstract
Approximately 70%-85% of breast cancers express androgen receptors (ARs). The role of AR in breast cancer pathogenesis is currently in exploration. Both androgens and anti-androgens have demonstrated variable inhibitory and stimulatory effects in AR-positive breast cancer depending on estrogen receptor and HER2 co-expression. Androgen signaling pathways interact with other critical cellular pathways, such as the PI3K/AKT/mTOR, Ras/Raf/MAPK/ERK, Wnt/β-catenin, and estrogen signaling pathways. Therapeutic exploitation of AR has been the crux of management of prostate cancer for decades. In recent years there has been increasing interest in AR as a novel therapeutic target in breast cancer. There have been many early phase clinical trials evaluating the safety and efficacy of various AR-targeted agents in breast cancer. Some of these studies have shown promising clinical benefits. Studies of biomarkers to identify the patients likely to benefit from AR-targeted therapies are currently in progress. Besides, AR expression may be an important prognostic and predictive marker for breast cancer, which needs to be defined better in future studies.
Collapse
Affiliation(s)
- Nusrat Jahan
- Division of Hematology-Oncology, Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4(th) St, Lubbock, Tx, 79430, USA.
| | - Catherine Jones
- Division of Hematology-Oncology, Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4(th) St, Lubbock, Tx, 79430, USA
| | - Rakhshanda Layeequr Rahman
- Department of Surgery, Texas Tech University Health Sciences Center, 3601 4(th)St, Lubbock, Tx, 79430, USA
| |
Collapse
|
5
|
Al-Othman N, Ahram M, Alqaraleh M. Role of androgen and microRNA in triple-negative breast cancer. Breast Dis 2020; 39:15-27. [PMID: 31839601 DOI: 10.3233/bd-190416] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Breast cancer (BC) is the most frequent type of malignancy affecting females worldwide. Molecular-based studies resulted in an identification of at least four subtypes of breast carcinoma, including luminal A and luminal B, Human growth factor receptor (HER-2)-enriched and triple-negative tumors (basal-like and normal breast-like). A proportion of BC cases are of the triple-negative breast cancer (TNBC) type. TNBC lacks the expression of estrogen receptor (ER), progesterone receptor (PR), and HER-2, and is known to express androgen receptor (AR) at considerable levels. AR has been shown to promote the progression of TNBC. However, the exact mechanisms have yet to be unraveled. One of these mechanisms could be through regulating the expression of microRNA (miRNA) molecules, which play an important regulatory role in BC through post-transcriptional gene silencing. Activation of AR controls the expression of miRNA molecules, which target selective mRNAs, consequently, affecting protein expression. In this review we attempt to elucidate the relations between AR and miRNA in TNBC.
Collapse
Affiliation(s)
- Nihad Al-Othman
- Division of Anatomy, Biochemistry and Genetic, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Mamoun Ahram
- Department of Physiology and Biochemistry, School of Medicine, The University of Jordan, Amman, Jordan
| | - Moath Alqaraleh
- Department of Biological Sciences, School of Science, The University of Jordan, Amman, Jordan
| |
Collapse
|
6
|
Verma P, Mittal P, Singh A, Singh IK. New Entrants into Clinical Trials for Targeted Therapy of Breast Cancer: An Insight. Anticancer Agents Med Chem 2020; 19:2156-2176. [PMID: 31656157 DOI: 10.2174/1871520619666191018172926] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 02/08/2023]
Abstract
Breast cancer is too complex with various different molecular alterations involved in its pathogenesis and progression. Over the decade, we have seen a surge in the development of drugs for bimolecular targets and for the signal transduction pathways involved in the treatment line of breast cancer. These drugs, either alone or in combination with conventional treatments like chemotherapy, hormone therapy and radiotherapy, will help oncologists to get a better insight and do the needful treatment. These novel therapies bring various challenges along with them, which include the dosage selection, patient selection, schedule of treatment and weighing of clinical benefits over side effects. In this review, we highlight the recently studied target molecules that have received indications in breast carcinoma, both in the localized and in an advanced state and about their inhibitors which are in clinical development which can give the immense potential to clinical care in the near future.
Collapse
Affiliation(s)
- Priyanka Verma
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi, 110019, India
| | - Pooja Mittal
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi, 110019, India
| | - Archana Singh
- Department of Botany, Hansraj College, University of Delhi, New Delhi, 110007, India.,Department of Molecular Ecology, Max-Planck Institute for Chemical Ecology, Hans-Knöll-Straße 8, D-07745 Jena, Germany
| | - Indrakant K Singh
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi, 110019, India.,Department of Molecular Ecology, Max-Planck Institute for Chemical Ecology, Hans-Knöll-Straße 8, D-07745 Jena, Germany
| |
Collapse
|
7
|
Evaluation of anti-tumour properties of two depsidones - Unguinol and Aspergillusidone D - in triple-negative MDA-MB-231 breast tumour cells. Toxicol Rep 2019; 6:1216-1222. [PMID: 31788435 PMCID: PMC6880138 DOI: 10.1016/j.toxrep.2019.10.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 11/21/2022] Open
Abstract
Unguinol and Aspergillusidone D inhibit cell viability at low μM concentrations. These depsidones increase the amount of apoptotic cells in the MDA-MB-231 cell line. Unguinol causes cell cycle arrest of MDA-MB-231 cells in the G2/M-phase. Apoptosis and cell cycle arrest were seen at clinically high concentrations (>60 μM).
There is an ongoing search for new compounds to lower the mortality and recurrence of breast cancer, especially triple-negative breast cancer. Naturally occurring depsidones, extracted from the fungus Aspergillus, are known for their wide range of biological activities such as cytotoxicity, aromatase inhibition, radical scavenging, and antioxidant properties. Research showed the potential of depsidones as a treatment option for hormone receptor-positive breast cancer treatment, yet its effects on hormone receptor-negative breast cancer are still unkown. This study, therefore, investigated the potential of two depsidones (Unguinol and Aspergillusidone D) to induce apoptosis, cell cycle arrest and cytotoxicity, and reduce cell proliferation in the triple-negative MDA-MB-231 breast cancer cell line. Results were compared with the effects of the cytostatic drug doxorubicin, antimitotic agent colchicine and endogenous hormones 17β-estradiol, testosterone and dihydrotestosterone. The cytostatic drugs and hormones affected the MDA-MB-231 cell line comparable to other studies, showing the usefulness of this model to study the effects of depsidones on a triple-negative breast cancer cell line. At sub μM levels, Unguinol and Aspergillusidone D did not influence cell proliferation, while cell viability was reduced at concentrations higher than 50 μM. Both depsidones induced apoptosis, albeit not statistically significantly. In addition, Unguinol induced cell cycle arrest in MDA-MB-231 cells at 100 μM. Our research shows the potential of two depsidones to reduce triple-negative breast cancer cell survival. Therefore, this group of compounds may be promising in the search for new cancer treatments, especially when looking at similar depsidones.
Collapse
|
8
|
Venema CM, Bense RD, Steenbruggen TG, Nienhuis HH, Qiu SQ, van Kruchten M, Brown M, Tamimi RM, Hospers GAP, Schröder CP, Fehrmann RSN, de Vries EGE. Consideration of breast cancer subtype in targeting the androgen receptor. Pharmacol Ther 2019; 200:135-147. [PMID: 31077689 DOI: 10.1016/j.pharmthera.2019.05.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/26/2019] [Indexed: 02/05/2023]
Abstract
The androgen receptor (AR) is a drug target in breast cancer, and AR-targeted therapies have induced tumor responses in breast cancer patients. In this review, we summarized the role of AR in breast cancer based on preclinical and clinical data. Response to AR-targeted therapies in unselected breast cancer populations is relatively low. Preclinical and clinical data show that AR antagonists might have a role in estrogen receptor (ER)-negative/AR-positive tumors. The prognostic value of AR for patients remains uncertain due to the use of various antibodies and cut-off values for immunohistochemical assessment. To get more insight into the role of AR in breast cancer, we additionally performed a retrospective pooled analysis to determine the prognostic value of the AR using mRNA profiles of 7270 primary breast tumors. Our analysis shows that a higher AR mRNA level is associated with improved disease outcome in patients with ER-positive/human epidermal growth factor receptor 2 (HER2)-negative tumors, but with worse disease outcome in HER2-positive subgroups. In conclusion, next to AR expression, incorporation of additional tumor characteristics will potentially make AR targeting a more valuable therapeutic strategy in breast cancer.
Collapse
Affiliation(s)
- Clasina M Venema
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rico D Bense
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Tessa G Steenbruggen
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hilde H Nienhuis
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Si-Qi Qiu
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Michel van Kruchten
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, United States
| | - Rulla M Tamimi
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Geke A P Hospers
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Carolina P Schröder
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rudolf S N Fehrmann
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
9
|
Morgan MM, Arendt LM, Alarid ET, Beebe DJ, Johnson BP. Mammary adipose stromal cells derived from obese women reduce sensitivity to the aromatase inhibitor anastrazole in an organotypic breast model. FASEB J 2019; 33:8623-8633. [PMID: 31002529 DOI: 10.1096/fj.201802347rrr] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aromatase inhibitors are the preferred treatment for certain women with estrogen receptor (ER)-positive breast cancer, but evidence suggests that women with obesity experience aromatase inhibitor resistance at higher rates. To compare how stromal cells derived from women who are lean or obese influence response to the aromatase inhibitor (anastrazole), we incorporated patient-derived stroma in a previously characterized MCF7-derived in vitro duct model. Coculture with adipose stromal cells enabled the metabolism of testosterone (T) to E2, which induced estrogen response element activity, epithelial proliferation, and hyperplasia in MCF7 cells. The effects of T were inhibited by the ER antagonist tamoxifen and aromatase inhibitor anastrazole and were increased by the aromatase inducer dexamethasone. Primary mammary adipose stromal cells derived from women with obesity displayed increased aromatase mRNA compared with lean controls. MCF7-derived ducts cocultured with obese stromal cells exhibited higher maximal aromatization-induced ER transactivation and reduced anastrazole sensitivity, a difference not seen in 2-dimensional coculture. Finally, tamoxifen was more effective than anastrazole at reducing aromatization-induced ER transactivation and proliferation. These findings suggest that patient-specific responses to hormone therapies can be modeled and studied organotypically in vitro and add to evidence advocating obesity as a parameter to consider when identifying treatments for patients with ER-positive breast cancer.-Morgan, M. M., Arendt, L. M., Alarid, E. T., Beebe, D. J., Johnson, B. P. Mammary adipose stromal cells derived from obese women reduce sensitivity to the aromatase inhibitor anastrazole in an organotypic breast model.
Collapse
Affiliation(s)
- Molly M Morgan
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lisa M Arendt
- Department of Comparative Biosciences, University Wisconsin-Madison, Madison, Wisconsin, USA.,Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Elaine T Alarid
- Department of Oncology, University Wisconsin-Madison, Madison, Wisconsin, USA.,Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David J Beebe
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Brian P Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
10
|
Depsidones inhibit aromatase activity and tumor cell proliferation in a co-culture of human primary breast adipose fibroblasts and T47D breast tumor cells. Toxicol Rep 2017; 4:165-171. [PMID: 28959637 PMCID: PMC5615129 DOI: 10.1016/j.toxrep.2017.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/13/2017] [Accepted: 03/17/2017] [Indexed: 01/15/2023] Open
Abstract
Depsidones are aromatase inhibitors in primary human breast adipose fibroblasts. Depsidones may have pharmacotherapeutical relevance for breast cancer treatment. Co-cultures of breast tumor and fibroblasts cells create a vivo realistic in vitro model for estrogen dependent breast cancer.
Naturally occurring depsidones from the marine fungus Aspergillus unguis are known to have substantial anti-cancer activity, but their mechanism of action remains elusive. The purpose of this study was to examine the anti-aromatase activity of two common depsidones, unguinol and aspergillusidone A, in a co-culture system of human primary breast adipose fibroblasts and hormonal responsive T47D breast tumor cells. Using this in vitro model it was shown that these depsidones inhibit the growth of T47D tumor cells most likely via inhibition of aromatase (CYP19) activity. The IC50 values of these depisidones were compared with the aromatase inhibitors letrozole and exemestane. Letrozole and exemestane had IC50 values of respectively, 0.19 and 0.14 μM, while those for Unguinol and Aspergillusidone A were respectively, 9.7 and 7.3 μM. Our results indicate that among the depsidones there maybe aromatase inhibitors with possible pharmacotherapeutical relevance.
Collapse
|
11
|
Androgen Receptor Expression in Thai Breast Cancer Patients. Med Sci (Basel) 2016; 4:medsci4030015. [PMID: 29083379 PMCID: PMC5635797 DOI: 10.3390/medsci4030015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/19/2016] [Accepted: 09/02/2016] [Indexed: 01/05/2023] Open
Abstract
The aim of this study was to investigate prevalence and related factors of androgen receptor (AR) expression in Thai breast cancer patients. A descriptive study was done in 95 patients, who were admitted to Charoenkrung Pracharak Hospital, Bangkok (2011-2013). Statistical relationships were examined between AR protein expression, tumor status, and patient characteristics. Compared with those from Western countries, ethnic Thai patients were younger at age of diagnosis and had a higher proliferative index (high Ki-67 expression), which indicates unfavorable prognosis. In addition, 91% of the Thai breast tumors that were positive for any of the following receptors, estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) also expressed the AR protein, while in triple negative breast tumors only 33% were AR positive. ER and PR expression was positively related with AR expression, while AR expression was inversely correlated to Ki-67 expression. AR status was strongly correlated with ER and PR status in Thai patients. There is an inverse relationship between Ki-67 and AR, which suggests that AR may be a prognostic factor for breast cancer.
Collapse
|
12
|
Marques R, Peres CG, Vaz CV, Gomes IM, Figueira MI, Cairrão E, Verde I, Maia CJ, Socorro S. 5α-Dihydrotestosterone regulates the expression of L-type calcium channels and calcium-binding protein regucalcin in human breast cancer cells with suppression of cell growth. Med Oncol 2015; 32:228. [PMID: 26255063 DOI: 10.1007/s12032-015-0676-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/03/2015] [Indexed: 12/20/2022]
Abstract
Androgens have been associated with the development of normal breast, and their role in mammary gland carcinogenesis has also been described. Several studies reported that androgens inhibit breast cancer cell growth, whereas others linked their action with the modulation of calcium (Ca(2+)) pumps, Ca(2+) channels and Ca(2+)-binding proteins. Also, it is known that deregulated Ca(2+) homeostasis has been implicated in the pathophysiology of breast. The L-type Ca(2+) channels (LTCCs) were found to be up-regulated in colon, colorectal and prostate cancer, but their presence in breast tissues remains uncharacterized. On the other hand, regucalcin (RGN) is a Ca(2+)-binding protein involved in the control of mammary gland cell proliferation, which has been identified as an androgen target gene in distinct tissues except breast. This study aimed to confirm the expression and activity of LTCCs in human breast cancer cells and investigate the effect of androgens in regulating the expression of α1C subunit (Cav1.2) of LTCCs and Ca(2+)-binding protein RGN. PCR, Western blot, immunofluorescence and electrophysiological experiments demonstrated the expression and activity of Cav1.2 subunit in MCF-7 cells. The MCF-7 cells were treated with 1, 10 or 100 nM of 5α-dihydrotestosterone (DHT) for 24-72 h. The obtained results showed that 1 nM DHT up-regulated the expression of Cav1.2 subunit while diminishing RGN protein levels, which was underpinned by reduced cell viability. These findings first confirmed the presence of LTCCs in breast cancer cells and opened new perspectives for the development of therapeutic approaches targeting Ca(2+) signaling.
Collapse
Affiliation(s)
- Ricardo Marques
- Faculdade de Ciências da Saúde, CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Safarpour D, Tavassoli FA. A targetable androgen receptor-positive breast cancer subtype hidden among the triple-negative cancers. Arch Pathol Lab Med 2014; 139:612-7. [PMID: 25310144 DOI: 10.5858/arpa.2014-0122-ra] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT Triple-negative breast cancer (TNBC) is a subgroup of breast cancers that by definition lack expression of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2). A diverse group of tumors, TNBC shares some morphologic and molecular features with basal-like breast cancer, a category of breast cancer defined by gene expression profiling. More likely to occur in young women and African Americans, TNBCs may exhibit aggressive behavior and are associated with poor prognosis despite their initial response to conventional chemotherapy. Because hormonal or HER2-targeted therapies are ineffective for these tumors, the main therapeutic option is systemic chemotherapy. Therefore, identification of new targets for therapy is urgently needed for this group. OBJECTIVE To review and present recent literature along with our own experience regarding the clinical and morphologic characteristics and the prevalence of androgen receptor (AR) expression in TNBC, and to discuss the potential use of AR as a therapeutic target for AR(+) TNBC. DATA SOURCES Data sources are published articles from peer-reviewed journals in PubMed (US National Library of Medicine). CONCLUSIONS AR is the most commonly expressed hormone receptor among all breast carcinomas, with a prevalence of 25% to 75% among TNBCs. Therefore, we strongly support the routine assessment of AR in TNBC, and preferably in all breast carcinomas.
Collapse
Affiliation(s)
- Damoun Safarpour
- From the Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | | |
Collapse
|
14
|
Chottanapund S, Van Duursen MBM, Navasumrit P, Hunsonti P, Timtavorn S, Ruchirawat M, Van den Berg M. Anti-aromatase effect of resveratrol and melatonin on hormonal positive breast cancer cells co-cultured with breast adipose fibroblasts. Toxicol In Vitro 2014; 28:1215-21. [PMID: 24929094 DOI: 10.1016/j.tiv.2014.05.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 05/27/2014] [Accepted: 05/29/2014] [Indexed: 12/29/2022]
Abstract
Targeting the estrogen pathway has been proven effective in the treatment for estrogen receptor positive breast cancer. There are currently two common groups of anti-estrogenic compounds used in the clinic; Selective Estrogen Receptor Modulators (SERMs, e.g. tamoxifen) and Selective Estrogen Enzyme Modulators (SEEMs e.g. letrozole). Among various naturally occurring, biologically active compounds, resveratrol and melatonin have been suggested to act as aromatase inhibitors, which make them potential candidates in hormonal treatment of breast cancer. Here we used a co-culture model in which we previously demonstrated that primary human breast adipose fibroblasts (BAFs) can convert testosterone to estradiol, which subsequently results in estrogen receptor-mediated breast cancer T47D cell proliferation. In the presence of testosterone in this model, we examined the effect of letrozole, resveratrol and melatonin on cell proliferation, estradiol (E2) production and gene expression of CYP19A1, pS2 and Ki-67. Both melatonin and resveratrol were found to be aromatase inhibitors in this co-culture system, albeit at different concentrations. Our co-culture model did not provide any indications that melatonin is also a selective estrogen receptor modulator. In the T47D-BAF co-culture, a melatonin concentration of 20 nM and resveratrol concentration of 20 μM have an aromatase inhibitory effect as potent as 20 nM letrozole, which is a clinically used anti-aromatase drug in breast cancer treatment. The SEEM mechanism of action of especially melatonin clearly offers potential advantages for breast cancer treatment.
Collapse
Affiliation(s)
- Suthat Chottanapund
- Division of Environmental Toxicology, Chulabhorn Graduate Institute, Bangkok, Thailand; Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, Thailand; Center of Excellence on Environmental Health, Toxicology and Management of Chemicals, Bangkok, Thailand; Bamrasnaradura Infectious Diseases Institute, Ministry of Public Health, Thailand.
| | - M B M Van Duursen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Panida Navasumrit
- Division of Environmental Toxicology, Chulabhorn Graduate Institute, Bangkok, Thailand; Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, Thailand; Center of Excellence on Environmental Health, Toxicology and Management of Chemicals, Bangkok, Thailand
| | - Potchanee Hunsonti
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, Thailand
| | - Supatchaya Timtavorn
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, Thailand
| | - Mathuros Ruchirawat
- Division of Environmental Toxicology, Chulabhorn Graduate Institute, Bangkok, Thailand; Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok, Thailand; Center of Excellence on Environmental Health, Toxicology and Management of Chemicals, Bangkok, Thailand
| | - Martin Van den Berg
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
15
|
Holdsworth-Carson SJ, Zaitseva M, Girling JE, Vollenhoven BJ, Rogers PAW. Common fibroid-associated genes are differentially expressed in phenotypically dissimilar cell populations isolated from within human fibroids and myometrium. Reproduction 2014; 147:683-92. [DOI: 10.1530/rep-13-0580] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Uterine fibroids are a prevalent gynaecological condition in reproductive-aged women and are the commonest reason for hysterectomy. The cellular composition of clonal fibroids are heterogeneous, with phenotypically dissimilar cells that include smooth muscle cells (SMC), vascular SMC (VSMC) and fibroblasts. The aim of our study was to investigate genes that are commonly differentially expressed between fibroid and myometrial whole tissues in phenotypically different sub-populations of cells isolated from fibroid and myometrium. Genes to be investigated by fluorescence-activated cell sorting, quantitative real-time PCR and immunocytochemistry include transforming growth factor β (TGFB) and retinoic acid (RA) signalling families and steroid hormone receptors. We hypothesised that each cell population isolated from fibroid and myometrium would differ in the expression of fibroid-associated genes. We demonstrated that phenotypically different cellular constituents of uterine fibroids differentially express cellular RA-binding protein 2 (CRABP2), progesterone receptor B (PRB) and TGFB receptor 2 mRNA in fibroid-derived cells of VSMC and SMC phenotype. CRABP2 mRNA was also differentially expressed in fibroblasts and VSMC sub-populations from within clonal fibroid tumours. We conclude that differential regulation of RA, TGFB and PR pathway transcription occurs in fibroid-associated SMC and -fibroblasts and that investigation of paracrine interactions between different cell types within the fibroid microenvironment provides an important new paradigm for understanding the pathophysiology of this common disease.
Collapse
|
16
|
Weigelt B, Ghajar CM, Bissell MJ. The need for complex 3D culture models to unravel novel pathways and identify accurate biomarkers in breast cancer. Adv Drug Deliv Rev 2014; 69-70:42-51. [PMID: 24412474 PMCID: PMC4186247 DOI: 10.1016/j.addr.2014.01.001] [Citation(s) in RCA: 212] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 12/27/2013] [Accepted: 01/03/2014] [Indexed: 12/11/2022]
Abstract
The recent cataloging of the genomic aberrations in breast cancer has revealed the diversity and complexity of the disease at the genetic level. To unravel the functional consequences of specific repertoires of mutations and copy number changes on signaling pathways in breast cancer, it is crucial to develop model systems that truly recapitulate the disease. Here we discuss the three-dimensional culture models currently being used or recently developed for the study of normal mammary epithelial cells and breast cancer, including primary tumors and dormancy. We discuss the insights gained from these models in regards to cell signaling and potential therapeutic strategies, and the challenges that need to be met for the generation of heterotypic breast cancer model systems that are amenable for high-throughput approaches.
Collapse
Affiliation(s)
- Britta Weigelt
- Department of Pathology, Memorial-Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| | - Cyrus M Ghajar
- Public Health Sciences Division/Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Mina J Bissell
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
17
|
Paz AC, Soleas J, Poon JC, Trieu D, Waddell TK, McGuigan AP. Challenges and Opportunities for Tissue-Engineering Polarized Epithelium. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:56-72. [DOI: 10.1089/ten.teb.2013.0144] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Ana C. Paz
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - John Soleas
- Latner Thoracic Surgery Research Laboratories, McEwen Centre for Regenerative Medicine, Toronto General Hospital, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - James C.H. Poon
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Latner Thoracic Surgery Research Laboratories, McEwen Centre for Regenerative Medicine, Toronto General Hospital, Toronto, ON, Canada
| | - Dennis Trieu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Latner Thoracic Surgery Research Laboratories, McEwen Centre for Regenerative Medicine, Toronto General Hospital, Toronto, ON, Canada
| | - Thomas K. Waddell
- Latner Thoracic Surgery Research Laboratories, McEwen Centre for Regenerative Medicine, Toronto General Hospital, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Alison P. McGuigan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|