35 results on '"hormone-dependent breast cancer"'
Search Results
2. Novel synthetic derivatives of cinnamic and p-coumaric acids with antiproliferative effect on breast MCF-7 tumor cells.
- Author
-
Munhoz Rodrigues, Débora, Bulhões Portapilla, Gisele, Soubihe de Sicco, Gabriel, Reis da Silva, Iris Fraioli, de Albuquerque, Sergio, Kenupp Bastos, Jairo, and Leiria Campo, Vanessa
- Subjects
CINNAMIC acid derivatives ,PROPOLIS ,CINNAMIC acid ,BREAST tumors ,AMINO acid derivatives ,CANCER cell growth ,NATURAL products - Abstract
p-Coumaric acid is derived from cinnamic acid and is one of the major compounds in the Brazilian green propolis extract. Studies have shown that both p-coumaric acid and cinnamic acid have promising antiproliferative effects. In this context, aiming to increase the complexity of these active natural products and their activities, we performed coupling reactions with propargylamine and benzylamine, as well as with threonine, phenylalanine and lysine amino acids, aiming to enhance their antiproliferative effects towards the hormone-dependent breast cancer MCF-7 cells. Overall, the p-coumaric acid coupling with L-threonine amino acid (compound 15) had the best selectivity index (SI = 5.1), with half-maximal inhibitory concentration of 39.6 ± 1 µM, showing a high selectivity against hormone-dependent breast cancer cell lines MCF-7 and low cytotoxicity against the normal breast cell lines MCF-10A. Thus, this new natural product derivative may represent a prototype for the future development of antiproliferative agents, especially against hormone-dependent breast cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
3. Efficacy and safety of adjuvant ovarian suppression with Buserelin-depo in premenopausal women with hormone-positive breast cancer
- Author
-
L. K. Ovchinnikova and A. D. Belko
- Subjects
hormone-dependent breast cancer ,drug ovarian suppression ,buserelin-depo ,Medicine - Abstract
Breast cancer (BC) ranks first in the morbidity pattern among women in Russia. Adjuvant endocrinotherapy is an important step in the complex treatment of premenopausal patients with hormone-positive early breast cancer. The drug ovarian suppression with GnRH-agonists have supplanted surgical castration and radiation-based treatment of the ovaries in patients with hormone-positive early breast cancer. Today, several drugs authorised for the treatment of breast cancer are used in clinical practice: goserelin, buserelin and triptorelin. Buserelin-depo is an effective method for achieving ovarian suppression. The results obtained do not differ from similar indicators obtained in using imported LHRH analogues.
- Published
- 2019
- Full Text
- View/download PDF
4. Nuclear PDCD4 Expression Defines a Subset of Luminal B-Like Breast Cancers with Good Prognosis.
- Author
-
Madera, Santiago, Chervo, María F., Chiauzzi, Violeta A., Pereyra, Matías G., Venturutti, Leandro, Izzo, Franco, Roldán Deamicis, Agustina, Guzman, Pablo, Dupont, Agustina, Roa, Juan Carlos, Cenciarini, Mauro E., Barchuk, Sabrina, Figurelli, Silvina, Lopez Della Vecchia, Daniel, Levit, Claudio, Lebersztein, Gabriel, Anfuso, Fabiana, Castiglioni, Teresa, Cortese, Eduardo, and Ares, Sandra
- Abstract
The hormone receptor-positive (estrogen and/or progesterone receptor (PR)-positive) and HER2-negative breast cancer (BC) subtype is a biologically heterogeneous entity that includes luminal A-like (LumA-like) and luminal B-like (LumB-like) subtypes. Decreased PR levels is a distinctive biological feature of LumB-like tumors. These tumors also show reduced sensitivity to endocrine therapies and poorer prognosis than LumA-like tumors. Identification of biomarkers to accurately predict disease relapse in these subtypes is crucial in order to select effective therapies. We identified the tumor suppressor PDCD4 (programmed cell death 4), located in the nucleus (NPDCD4), as an independent prognostic factor of good clinical outcome in LumA-like and LumB-like subtypes. NPDCD4-positive LumB-like tumors presented overall and disease-free survival rates comparable to those of NPDCD4-positive LumA-like tumors, indicating that NPDCD4 improves the outcome of LumB-like patients. In contrast, NPDCD4 loss increased the risk of disease recurrence and death in LumB-like compared with LumA-like tumors. This, along with our results showing that LumB-like tumors present lower NPDCD4 positivity than LumA-like tumors, suggests that NPDCD4 loss contributes to endocrine therapy resistance in LumB-like BCs. We also revealed that PR induces PDCD4 transcription in LumB-like BC, providing a mechanistic explanation to the low PDCD4 levels in LumB-like BCs lacking PR. Finally, PDCD4 silencing enhanced BC cell survival in a patient-derived explant model of LumB-like disease. Our discoveries highlight NPDCD4 as a novel biomarker in LumA- and LumB-like subtypes, which could be included in the panel of immunohistochemical markers used in the clinic to accurately predict the prognosis of LumB-like tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
5. Ex vivo interaction between blood components and hormone-dependent breast cancer cells induces alterations associated with epithelial-mesenchymal transition and thrombosis.
- Author
-
Augustine, T. N., Pather, K., Mak, D., Klonaros, D., Xulu, K., Dix-Peek, T., Duarte, R., and van der Spuy, W. J.
- Subjects
- *
EPITHELIAL-mesenchymal transition , *TRANSFORMING growth factors-beta , *BREAST cancer , *CANCER cell culture , *CANCER cells , *FIBRIN - Abstract
The prevalence of breast cancer is steadily increasing with metastasis and thromboembolic complications identified as the most common causes of death. The acquisition of an aggressive phenotype by hormone-dependent breast cancers is mediated by Transforming Growth Factor Beta 1 (TGF-β1) expression and is associated with epithelial-mesenchymal transition (EMT) and, potentially, increased propensity for thrombosis. We investigated this phenomenon by assessing the effect of platelet-rich plasma (PRP) and whole blood (WB) on parameters of EMT and hypercoagulation in vitro. MCF-7 breast cancer cells were cultured under standard conditions, followed by co-culture with PRP or WB. Cells were processed for real-time PCR (TGF-β1 and vimentin), electron microscopy or immunocytochemistry (TGF-β1). Micrographs were qualitatively assessed, and real-time PCR data analyzed with PAST Statistical Software. The addition of blood components heightened TGF-β1 immunolocalization and significantly increased corresponding gene expression. Both PRP and WB significantly increased vimentin expression and induced a shape change from a typical epithelial phenotype to a spindle-shape morphology, indicative of EMT. Fibrin fiber, network and plaque formation indicated a hypercoagulatory environment. The results thus show that in preparation for hematogenous metastasis, hormone-dependent breast cancer cells assume an aggressive phenotype associated with EMT, simultaneously increasing the propensity for the formation of thrombo-emboli. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
6. Immunohistochemical evaluation of insulin-like growth factor receptor 1 in breast cancer
- Author
-
Batinić-Škipina Danijela, Marić Radmil, Tadić-Latinović Ljiljana, Erić Dražan, and Lalović Nenad
- Subjects
insulin-like growth factor 1 receptor (IGF-1R) ,hormone-dependent breast cancer ,HER-2 ,Medicine - Abstract
Introduction/Objective. Activation of insulin-like growth factor receptor (IGF-1R) results in cell transition from growth phase to synthesis phase of cell cycle. Breast cancer is categorized into prognostic and therapeutic subtypes based upon hormone receptor, estrogen receptor (ER), and progesterone receptor (PR) expression and human epidermal growth factor receptor 2 (HER-2) expression. The objective of this study was to examine the expression of IGF-1R in а specific subtype invasive breast cancer and its correlation with basic histopathological and immunohistochemical prognostic parameters. Methods. Formalin-fixed paraffin-embedded tumor samples were obtained from 129 female patients with invasive breast cancer (I–III disease stage) with the follow-up ranging 36–108 months (average 48 months). For immunohistochemical staining, we used monoclonal antibodies for ER, PR, IGF-1R, and polyclonal antibody for HER-2. Results. IGF-1R inversely correlated with tumor stage (p = 0.017), tumor grade (p = 0.001), HER-2 (p = 0.003), whereas significant positive correlation was found with multifocality/multicentricity of breast cancer (p = 0.036), ER (p = 0.001) and PR (p = 0.0001) expression. Cox-regression analysis for relapse-free survival (RFS) showed that disease stage (p = 0.039) and HER-2 (p = 0.033) were independent prognostic factors. IGF-1R did not predict clinical outcome in patients with breast cancer (p = 0.488, Kaplan–Meier test for RFS). Conclusion. Patients with low stage and grade hormone-dependent breast cancer had a significantly higher IGF-1R expression than patients with triple negative or HER-2 overexpressed cancer. The present findings also highlight that IGF-1R expression in multicentric/multifocal breast cancer supports the key roles in tumor initiation.
- Published
- 2018
- Full Text
- View/download PDF
7. The balance of estrogen metabolites in breast cancer and the ways of its correction
- Author
-
L. A. Ashrafyan, N. A. Babaeva, I. B. Antonova, O. A. Ovchinnikova, O. I. Aleshikova, T. A. Motskobili, and I. N. Kuznetsov
- Subjects
breast cancer ,estrogen metabolites ,2-hydroxyestrone ,16α-hydroxyestrone ,indinol ,indinol forto ,hormone-dependent breast cancer ,hormone-independent breast cancer ,targeted therapy ,Gynecology and obstetrics ,RG1-991 - Abstract
Background. A clear relationship is now found between the activity of estrogen metabolites and the development of tumors in estrogen-dependent tissues. Many international and Russian studies could identify a number of compounds involved in the regulation of estrogen metabolites. Indole-3-carbinol is one of these compounds that correct a ratio of 2-hydroxyestrone (2-OHE1) to 16-OHE1. It is a phytonutrient that is contained in Cruciferous vegetables and has antitumor activity. Our investigations used highly purified indole-3-carbinol (Indinol). The ability to identify the role of metabolic syndrome, by applying the current methodological approaches, allowed us to study the effect of highly purified indole-3-carbinol on the level of expression of estrogen metabolites and to attempt to expand views on procedures to prevent and treat breast tumors. Materials and methods. A total of 136 women were comprehensively examined; of them 44 patients formed a group of those with morphologically verified hormone-independent breast cancer (BC) and 42 patients were a group of those with hormone-dependent BC. A control group included 50 women without signs of breast disease. In all the patients, body mass index was calculated; the ratio of urinary estrogen metabolites (2-ОНЕ1/16α-ОНЕ1) was quantified; and biopsy specimens and operative material were histologically and immunohistologically studied. Results. The BC patients were noted to have the high expression of 16α-OHE1 and the low values of 2-OHE1 (ratio, 0.42) as compared to the control group (2.09), which was an important component of metabolic syndrome. The BC patients had the high level of concomitant endocrine metabolic disturbances with the high body mass index, which reflected the clinical manifestation of metabolic syndrome. Indinol used to treat both hormone-dependent and hormone-independent BC showed an antimetabolic effect that was most pronounced 6 months later. Conclusion. Thus, early correction of metabolic disturbances with antimetabolites is one of the important, pathogenetically sound areas in the prevention of BC.
- Published
- 2015
- Full Text
- View/download PDF
8. Incidence of comorbidities in women with breast cancer treated with tamoxifen or an aromatase inhibitor: an Australian population-based cohort study.
- Author
-
Huah Shin Ng, Koczwara, Bogda, Roder, David, Niyonsenga, Theo, and Vitry, Agnes
- Subjects
TAMOXIFEN ,AROMATASE inhibitors ,BREAST tumors ,LONGITUDINAL method ,RESEARCH methodology ,RESEARCH funding ,TIME ,COMORBIDITY ,DISEASE incidence ,PROPORTIONAL hazards models ,RETROSPECTIVE studies ,THERAPEUTICS - Abstract
Background: The development of comorbidities has become increasingly relevant with longer-term cancer survival. Objective: To assess the pattern of comorbidities among Australian women with breast cancer treated with tamoxifen or an aromatase inhibitor. Design: Retrospective cohort study using Pharmaceutical Benefits Scheme (PBS) data (10% sample) from January 2003 to December 2014. Dispensing claims data were used to identify comorbidities and classified with the Rx-Risk-V model. The breast cancer cohort had tamoxifen or an aromatase inhibitor dispensed between 2004 and 2011 with no switching between types of endocrine therapy. Comparisons were made between the breast cancer cohort and specific control groups (age- and sex-matched at 1:10 ratio without any dispensing of anti-neoplastic agents during the study period) for the development of five individual comorbidities over time using Cox regression models. Results: Women treated with tamoxifen had a higher incidence of cardiovascular conditions, diabetes, and pain or pain-inflammation, but a lower incidence of hyperlipidaemia compared with non-cancer control groups, as indicated by PBS data. Women treated with aromatase inhibitors were more likely to develop cardiovascular conditions, osteoporosis, and pain or pain-inflammation compared with non-cancer control groups. The risks of hyperlipidaemia and osteoporosis were significantly lower among tamoxifen users compared with aromatase inhibitor users. Conclusion: Women with hormone-dependent breast cancer treated with an endocrine therapy had a higher risk of developing specified comorbid conditions than women without cancer, with different comorbidity profiles for those on tamoxifen versus aromatase inhibitors. Further research into the causes and mechanism of development and management of comorbidities after cancer is needed. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
9. Novel Selective Estrogen Receptor Ligand Conjugates Incorporating Endoxifen-Combretastatin and Cyclofenil-Combretastatin Hybrid Scaffolds: Synthesis and Biochemical Evaluation.
- Author
-
Kelly, Patrick M., Keely, Niall O., Bright, Sandra A., Yassin, Bassem, Ana, Gloria, Fayne, Darren, Zisterer, Daniela M., and Meegan, Mary J.
- Subjects
- *
SELECTIVE estrogen receptor modulators , *NUCLEAR receptors (Biochemistry) , *LIGANDS (Biochemistry) , *BREAST cancer treatment , *OSTEOPOROSIS treatment - Abstract
Nuclear receptors such as the estrogen receptors (ERα and ERβ) modulate the effects of the estrogen hormones and are important targets for design of innovative chemotherapeutic agents for diseases such as breast cancer and osteoporosis. Conjugate and bifunctional compounds which incorporate an ER ligand offer a useful method of delivering cytotoxic drugs to tissue sites such as breast cancers which express ERs. A series of novel conjugate molecules incorporating both the ER ligands endoxifen and cyclofenil-endoxifen hybrids covalently linked to the antimitotic and tubulin targeting agent combretastatin A-4 were synthesised and evaluated as ER ligands. A number of these compounds demonstrated pro-apoptotic effects, with potent antiproliferative activity in ER-positive MCF-7 breast cancer cell lines and low cytotoxicity. These conjugates displayed binding affinity towards ERα and ERβ isoforms at nanomolar concentrations e.g., the cyclofenil-amide compound 13e is a promising lead compound of a clinically relevant ER conjugate with IC50 in MCF-7 cells of 187 nM, and binding affinity to ERα (IC50 = 19 nM) and ERβ (IC50 = 229 nM) while the endoxifen conjugate 16b demonstrates antiproliferative activity in MCF-7 cells (IC50 = 5.7 nM) and binding affinity to ERα (IC50 = 15 nM) and ERβ (IC50 = 115 nM). The ER binding effects are rationalised in a molecular modelling study in which the disruption of the ER helix-12 in the presence of compounds 11e, 13e and 16b is presented These conjugate compounds have potential application for further development as antineoplastic agents in the treatment of ER positive breast cancers [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
10. Novel synthetic derivatives of cinnamic and p -coumaric acids with antiproliferative effect on breast MCF-7 tumor cells.
- Author
-
Rodrigues DM, Portapilla GB, de Sicco GS, da Silva IFR, de Albuquerque S, Bastos JK, and Campo VL
- Subjects
- Humans, Female, Coumaric Acids pharmacology, MCF-7 Cells, Hormones pharmacology, Hormones therapeutic use, Cell Proliferation, Cell Line, Tumor, Antineoplastic Agents chemistry, Breast Neoplasms drug therapy
- Abstract
p -Coumaric acid is derived from cinnamic acid and is one of the major compounds in the Brazilian green propolis extract. Studies have shown that both p -coumaric acid and cinnamic acid have promising antiproliferative effects. In this context, aiming to increase the complexity of these active natural products and their activities, we performed coupling reactions with propargylamine and benzylamine, as well as with threonine, phenylalanine and lysine amino acids, aiming to enhance their antiproliferative effects towards the hormone-dependent breast cancer MCF-7 cells. Overall, the p -coumaric acid coupling with L -threonine amino acid (compound 15 ) had the best selectivity index (SI = 5.1), with half-maximal inhibitory concentration of 39.6 ± 1 μM, showing a high selectivity against hormone-dependent breast cancer cell lines MCF-7 and low cytotoxicity against the normal breast cell lines MCF-10A. Thus, this new natural product derivative may represent a prototype for the future development of antiproliferative agents, especially against hormone-dependent breast cancer.
- Published
- 2023
- Full Text
- View/download PDF
11. Unravelling exemestane: From biology to clinical prospects.
- Author
-
Sobral, Ana Filipa, Amaral, Cristina, Correia-da-Silva, Georgina, and Teixeira, Natércia
- Subjects
- *
EXEMESTANE , *HORMONE receptor positive breast cancer , *AROMATASE inhibitors , *ANTINEOPLASTIC agents , *CANCER invasiveness , *CANCER treatment , *PREVENTION , *THERAPEUTICS - Abstract
Aromatase inhibitors (AIs) are anti-tumor agents used in clinic to treat hormone-dependent breast cancer. AIs block estrogens biosynthesis by inhibiting the enzyme aromatase, preventing tumor progression. Exemestane, a third-generation steroidal AI, belongs to this class of drugs and is currently used in clinic to treat postmenopausal women, due to its high efficacy and good tolerability. Here, its pharmacological and biological aspects as well as its clinical applications and comparison to other endocrine therapeutic agents, are reviewed. It is also focused the benefits and risks of exemestane, drawbacks to be overcome and aspects to be explored. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
12. Molecular docking, QSAR, pharmacophore modeling, and dynamics studies of some chromone derivatives for the discovery of anti-breast cancer agents against hormone-dependent breast cancer.
- Author
-
Arvindekar SA, Mohole S, Patil A, Mane P, Arvindekar A, Mali SN, Thorat B, Rawat R, and Sharma S
- Subjects
- Female, Humans, Molecular Docking Simulation, Quantitative Structure-Activity Relationship, Pharmacophore, Chromones pharmacology, MCF-7 Cells, Hormones pharmacology, Cell Proliferation, Molecular Structure, Drug Screening Assays, Antitumor, Antineoplastic Agents chemistry, Neoplasms
- Abstract
In search of new anti-breast cancer agents, the present study envisaged the design and synthesis of a series of benzopyran-chalcones. All the synthesized compounds were assayed for their in-vitro anticancer activity against ER + MCF-7 and triple-negative MDA-MB-231 breast cancer cell lines using SRB assay. The synthesized compounds were found active against ER + MCF-7 cell lines. Based on the in-vitro data, in-silico analysis was performed using hormone-dependent breast cancer targets such as hER-α and aromatase because the compounds showed activity against MCF-7 cells and none was active against MDA-MB-231. The in-silico results supported the in-vitro anticancer activity suggesting the affinity of compounds toward hormone-dependant breast cancer. Compounds 4A1 to 4A3 were found to be most cytotoxic to MCF-7 cells with IC
50 values of 31.87, 22.95, and 20.34 μg/ml, respectively (Doxorubicin IC50 : <10 μg/ml). In addition, they showed the interactions with the amino acid residues of a binding cavity of an hER-α. Furthermore, quantitative structure-activity relationship (QSAR) studies were performed to reveal the vital structural features required for anticancer activity against breast cancer. Molecular dynamic simulation studies of hER-α and 4A3 in comparison with the raloxifene complex ensure the appropriate refinement of compounds in the dynamic system. Additionally, a generated pharmacophore model explored the essential pharmacophoric features of the synthesized scaffolds with respect to clinically used drug molecules for optimal hormone-dependant anti-breast cancer activity.Communicated by Ramaswamy H. Sarma.- Published
- 2023
- Full Text
- View/download PDF
13. Design, Synthesis and Biochemical Evaluation of Novel Selective Estrogen Receptor Ligand Conjugates Incorporating an Endoxifen-Combretastatin Hybrid Scaffold
- Author
-
Niall O. Keely, Miriam Carr, Bassem Yassin, Gloria Ana, David G. Lloyd, Daniela Zisterer, and Mary J. Meegan
- Subjects
estrogen receptor ligands ,selective estrogen receptor modulators ,tumour targeting ,conjugates ,tamoxifen ,endoxifen ,hormone-dependent breast cancer ,Biology (General) ,QH301-705.5 - Abstract
Nuclear-receptors are often overexpressed in tumours and can thereby be used as targets when designing novel selective chemotherapeutic agents. To date, many conjugates incorporating an estrogen receptor (ER) ligand have been synthesised in order to direct chemical agents to tissue sites containing ERs. A series of ER ligand conjugates were synthesised incorporating an antagonistic ER ligand scaffold based on endoxifen, covalently-bound via an amide linkage to a variety of combretastatin-based analogues, which may act as antimitotic agents. These novel endoxifen-combretastatin hybrid scaffold analogues were biochemically evaluated in order to determine their antiproliferative and cytotoxicity effects in both the ER-positive MCF-7 and the ER-negative MDA-MB-231 human breast cancer cell lines. ER competitive binding assays were carried out to assess the binding affinity of the lead conjugate 28 towards both the ERα and ERβ isoforms. In results from the NCI 60-cell line screen, the lead conjugate 28 displayed potent and highly selective antiproliferative activity towards the MCF-7 human cancer cell line (IC50 = 5 nM). In the ER-binding assays, the lead conjugate 28 demonstrated potent ER competitive binding in ERα (IC50 value: 0.9 nM) and ERβ (IC50 value: 4.7 nM). Preliminary biochemical results also demonstrate that the lead conjugate 28 may exhibit pure antagonism. This series makes an important addition to the class of ER antagonists and may have potential applications in anticancer therapy.
- Published
- 2016
- Full Text
- View/download PDF
14. Exemestane metabolites: Synthesis, stereochemical elucidation, biochemical activity and anti-proliferative effects in a hormone-dependent breast cancer cell line.
- Author
-
Varela, Carla L., Amaral, Cristina, Tavares da Silva, Elisiário, Lopes, Andreia, Correia-da-Silva, Georgina, Carvalho, Rui A., Costa, Saul C.P., Roleira, Fernanda M.F., and Teixeira, Natércia
- Subjects
- *
BREAST cancer treatment , *EXEMESTANE , *CHEMICAL synthesis , *METABOLITES , *STEREOCHEMISTRY , *ANTINEOPLASTIC agents , *CANCER hormone therapy - Abstract
Exemestane is a third-generation steroidal aromatase inhibitor that has been used in clinic for hormone-dependent breast cancer treatment in post-menopausal women. It is known that exemestane undergoes a complex metabolization, giving rise to some already identified metabolites, the 17β-hydroxy-6-methylenandrosta-1,4-dien-3-one (17- βHE ) and the 6-(hydroxymethyl)androsta-1,4,6-triene-3,17-dione ( 6-HME ). In this study, four metabolites of exemestane have been analyzed, three of them were synthesized (6β-spirooxiranandrosta-1,4-diene-3,17-dione ( 2 ), 1α,2α-epoxy-6-methylenandrost-4-ene-3,17-dione ( 3 ) and 17-βHE ( 4 )) while one was acquired, the 6-HME ( 6 ). The stereochemistry of the epoxide group of 2 and 3 has been unequivocally elucidated for the first time on the basis of NOESY experiments. New structure–activity relationships (SAR) have been established through the observation that the substitution of the double bonds by epoxide groups led to less potent derivatives in microsomes. However, the reduction of the C-17 carbonyl group to a hydroxyl group originating 17-βHE ( 4 ) resulted in a significant increase in activity in MCF-7aro cells when compared to exemestane (IC 50 0.25 μM vs 0.90 μM, respectively). All the studied metabolites reduced MCF-7aro cells viability in a dose and time-dependent manner, and metabolite 3 was the most potent one. Altogether our results showed that not only exemestane but also its main metabolites are potent aromatase inhibitors and reduce breast cancer cells viability. This suggests that exemestane efficacy may also be due to the active metabolites that result from its metabolic transformation. Our results emphasize the importance of performing further studies to expand our understanding of exemestane actions in breast cancer cells. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
15. Steroidal aromatase inhibitors inhibit growth of hormone-dependent breast cancer cells by inducing cell cycle arrest and apoptosis.
- Author
-
Amaral, Cristina, Varela, Carla, Borges, Margarida, Tavares da Silva, Elisiário, Roleira, Fernanda, Correia-da-Silva, Georgina, and Teixeira, Natércia
- Abstract
Different hormonal therapies are used for estrogen receptor positive (ER) breast cancers, being the third-generation of aromatase inhibitors (AIs), an effective alternative to the classical tamoxifen. AIs inhibit the enzyme aromatase, which is responsible for catalyzing the conversion of androgens to estrogens. In this study, it was evaluated the effects of several steroidal AIs, namely 3β-hydroxyandrost-4-en-17-one ( 1), androst-4-en-17-one ( 12), 4α,5α-epoxyandrostan-17-one ( 13a) and 5α-androst-2-en-17-one ( 16), on cell proliferation, cell cycle progression and cell death in an ER aromatase-overexpressing human breast cancer cell line (MCF-7aro). All AIs induced a decrease in cell proliferation and these anti-proliferative effects were due to a disruption in cell cycle progression and cell death, by apoptosis. AIs 1 and 16 caused cell cycle arrest in G/G, while AIs 12 and 13a induced an arrest in G/M. Moreover, it was observed that these AIs induced apoptosis by different pathways, since AIs 1, 12 and 13a activated the apoptotic mitochondrial pathway, while AI 16 induced apoptosis through activation of caspase-8. These results are important for the elucidation of the cellular effects of steroidal AIs on breast cancer cells and will also highlight the importance of AIs as inducers of apoptosis in hormone-dependent breast cancers. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
16. Local adipocytes enable estrogen-dependent breast cancer growth: Role of leptin and aromatase.
- Author
-
Liu, Enbo, Samad, Fahumiya, and Mueller, Barbara M
- Subjects
- *
FAT cells , *ESTROGEN , *BREAST cancer , *LEPTIN , *AROMATASE , *CANCER invasiveness - Abstract
The importance of the microenvironment in breast cancer growth and progression is becoming increasingly clear. Adipocytes are abundant in the mammary microenvironment, and recent studies show that adipocytes produce endocrine, inflammatory, and angiogenic factors that have tremendous potential to affect adjacent breast cancer cells. Yet, the extent to which local adipocyte function contributes to the pathogenesis of breast cancer is largely unexplored. Here we describe a unique animal model to study interactions between adipocytes and breast cancer cells in the tumor microenvironment. Our results suggest that local interactions between adipocytes and tumor cells are sufficient to promote the growth of hormone-dependent breast cancer. We also demonstrate that leptin signaling in adipocytes induces aromatase expression, expected to result in higher estrogen in the microenvironment thus enabling mammary tumorigenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
17. Effects of steroidal aromatase inhibitors on sensitive and resistant breast cancer cells: Aromatase inhibition and autophagy
- Author
-
Amaral, Cristina, Varela, Carla, Azevedo, Margarida, da Silva, Elisiário Tavares, Roleira, Fernanda M.F., Chen, Shiuan, Correia-da-Silva, Georgina, and Teixeira, Natércia
- Subjects
- *
AROMATASE inhibitors , *BREAST cancer treatment , *AUTOPHAGY , *DRUG resistance in cancer cells , *ESTROGEN receptors , *TAMOXIFEN , *STEROID synthesis , *ANDROSTENEDIONE - Abstract
Abstract: Several therapeutic approaches are used in estrogen receptor positive (ER+) breast cancers, being one of them the use of aromatase inhibitors (AIs). Although AIs demonstrate higher efficacy than tamoxifen, they can also exhibit de novo or acquired resistance after prolonged treatment. Recently, we have described the synthesis and biochemical evaluation of four steroidal AIs, 3β-hydroxyandrost-4-en-17-one (1), androst-4-en-17-one (12), 4α,5α-epoxyandrostan-17-one (13a) and 5α-androst-2-en-17-one (16), obtained from modifications in the A-ring of the aromatase substrate, androstenedione. In this study, it was investigated the biological effects of these AIs in different breast cancer cell lines, an ER+ aromatase-overexpressing human breast cancer cell line (MCF-7aro cells), an estrogen-receptor negative (ER−) human breast cancer cell line (SK-BR-3 cells), and a late stage of acquired resistance cell line (LTEDaro cells). The effects of an autophagic inhibitor (3-methyladenine) plus AIs 1, 12, 13a or exemestane in LTEDaro cells were also studied to understand the involvement of autophagy in AI acquired resistance. Our results showed that these steroids inhibit aromatase of MCF-7aro cells and decrease cell viability in a dose- and time-dependent manner. The new AI 1 is the most potent inhibitor, although the AI 12 demonstrates to be the most effective in decreasing cell viability. Besides, and in advantage over exemestane, AIs 12 and 13a also reduced LTEDaro cells viability. The use of the autophagic inhibitor allowed AIs to diminish viability of LTEDaro cells, presenting a similar behavior to the sensitive cells. Thus, inhibition of autophagy may sensitize hormone-resistant cancer cells to anti-estrogen therapies. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
18. Development of steroid sulfatase inhibitors
- Author
-
Woo, L.W. Lawrence, Purohit, Atul, and Potter, Barry V.L.
- Subjects
- *
ARYLSULFATASES , *HYDROLYSIS , *ESTROGEN , *HORMONE-dependent tumors , *BREAST cancer , *CLINICAL trials , *AROMATASE , *ENZYME inhibitors - Abstract
Abstract: Hydrolysis of biologically inactive steroid sulfates to unconjugated steroids by steroid sulfatase (STS) is strongly implicated in rendering estrogenic stimulation to hormone-dependent cancers such as those of the breast. Considerable progress has been made in the past two decades with regard to the discovery, design and development of STS inhibitors. We outline historical aspects of their development, cumulating in the discovery of the first clinical trial candidate STX64 (BN83495, Irosustat) and other sulfamate-based inhibitors. The development of reversible STS inhibitors and the design of dual inhibitors of both aromatase and STS is also discussed. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
19. Biochemical and biological evaluation of novel potent coumarin inhibitor of 17β-HSD type 1
- Author
-
Starčević, Š., Kocbek, P., Hribar, G., Lanišnik Rižner, T., and Gobec, S.
- Subjects
- *
ALCOHOL dehydrogenase , *COUMARINS , *ENZYME inhibitors , *NAD+ synthase , *CATALYSIS , *ESTROGEN receptors , *ESTRONE , *ESTRADIOL - Abstract
Abstract: Human 17β-hydroxysteroid dehydrogenase (17β-HSD) type 1 is an enzyme that acts at the pre-receptor level. It catalyzes the NADPH-dependent reduction of the weak estrogen estrone into the most potent estrogen 17β-estradiol, which exerts proliferative effects via estrogen receptors. Overexpression of 17β-HSD type 1 in estrogen-responsive tissues is related to the development of hormone-dependent diseases, such as breast cancer and endometriosis. 17β-HSD type 1 thus represents an attractive target for development of new drugs. Recently, we discovered that substituted coumarin derivatives potently and selectively inhibit 17β-HSD type 1. In the present study, we have performed additional biochemical and biological evaluation of the most promising coumarin derivative. First, we used an efficient method for isolation and purification of the active, soluble recombinant human 17β-HSD type 1 from Escherichia coli. This 17β-HSD type 1 showed a specific activity of 0.64±0.08μmolmin−1 mg−1 for estrone reduction in the presence of NADPH at pH 6.5, and a K m of 62nM for estrone. Next, we evaluated the best of the coumarin-derivative inhibitors, showing its reversible and competitive inhibition of 17β-HSD type 1 reductive activity with a K i of 53nM. We confirmed that this coumarin inhibitor acts not only in a cell-free assay, but also decreases endogenous 17β-HSD type 1 activity in human T-47D breast cancer cells. This inhibitor also reduced estrone dependent growth of T-47D cells after 48h of incubation. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
20. Design and synthesis of substrate mimetics based on an indole scaffold: potential inhibitors of 17β-HSD type 1.
- Author
-
Starčević, Štefan, Božnar, Polona, Turk, Samo, Gobec, Stanislav, and Rižner, Tea Lanišnik
- Subjects
- *
DEHYDROGENASES , *ALCOHOL dehydrogenase , *ESTROGEN , *STEROID hormones , *BREAST cancer , *ESCHERICHIA coli , *COLIFORMS , *ESTRONE , *PHENOL - Abstract
Background: Human 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1) acts at a pre-receptor level. It catalyzes NADPH-dependent reduction of the weak estrogen estrone into the most potent estrogen estradiol, which exerts its proliferative effects via estrogen receptors. Overexpression of 17β-HSD1 in estrogen-responsive tissues is related to the development of hormone-dependent diseases, such as breast cancer and endometriosis. 17β-HSD1 thus represents an attractive target for development of new drugs. Methods: We designed and synthesized a series of 3-, 5- and 6-phenyl indole derivatives as mimetics of the steroid substrate estrone. All of these compounds were evaluated for inhibition of recombinant human 17β-HSD1 from Escherichia coli, at concentrations of 0.6 μM and 6.0 μM. Results: Among 14 indole derivatives, compound 9 was an initial hit inhibitor of 17β-HSD1, with moderate inhibition (64% at 6 μM). Molecular docking into the crystal structure of 17β-HSD1 (1A27) revealed that this 5-phenyl indole derivative binds to 17β-HSD1 similarly to co-crystalized E2. Compound 9 forms two H-bonds with 17β-HSD1: one between the indole nitrogen and His222, and the second between the phenolic OH group and catalytic Tyr155. Conclusions: The indole scaffold is one of the possible starting points for the design of substrate mimetics of the steroid substrate estrone. Our study shows that these 6- and, especially, 5-phenol indole derivatives can act as moderate inhibitors of 17β-HSD1. Based on inhibition assays and docking simulations, we can infer further improvements of the 5-phenol indole derivatives that might result in better inhibition profiles. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
21. Immunohistochemical evaluation of insulin-like growth factor receptor 1 in breast cancer
- Author
-
Nenad Lalovic, Ljiljana Tadic-Latinovic, Radmil Marić, Drazan Eric, and Danijela Batinic-Skipina
- Subjects
Oncology ,030213 general clinical medicine ,medicine.medical_specialty ,business.industry ,lcsh:R ,lcsh:Medicine ,General Medicine ,Insulin-Like Growth Factor Receptor ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,HER-2 ,030220 oncology & carcinogenesis ,Internal medicine ,insulin-like growth factor 1 receptor (IGF-1R) ,medicine ,Immunohistochemistry ,hormone-dependent breast cancer ,business - Abstract
Introduction/Objective. Activation of insulin-like growth factor receptor (IGF-1R) results in cell transition from growth phase to synthesis phase of cell cycle. Breast cancer is categorized into prognostic and therapeutic subtypes based upon hormone receptor, estrogen receptor (ER), and progesterone receptor (PR) expression and human epidermal growth factor receptor 2 (HER-2) expression. The objective of this study was to examine the expression of IGF-1R in ? specific subtype invasive breast cancer and its correlation with basic histopathological and immunohistochemical prognostic parameters. Methods. Formalin-fixed paraffin-embedded tumor samples were obtained from 129 female patients with invasive breast cancer (I?III disease stage) with the follow-up ranging 36?108 months (average 48 months). For immunohistochemical staining, we used monoclonal antibodies for ER, PR, IGF-1R, and polyclonal antibody for HER-2. Results. IGF-1R inversely correlated with tumor stage (p = 0.017), tumor grade (p = 0.001), HER-2 (p = 0.003), whereas significant positive correlation was found with multifocality/multicentricity of breast cancer (p = 0.036), ER (p = 0.001) and PR (p = 0.0001) expression. Cox-regression analysis for relapse-free survival (RFS) showed that disease stage (p = 0.039) and HER-2 (p = 0.033) were independent prognostic factors. IGF-1R did not predict clinical outcome in patients with breast cancer (p = 0.488, Kaplan?Meier test for RFS). Conclusion. Patients with low stage and grade hormone-dependent breast cancer had a significantly higher IGF-1R expression than patients with triple negative or HER-2 overexpressed cancer. The present findings also highlight that IGF-1R expression in multicentric/multifocal breast cancer supports the key roles in tumor initiation.
- Published
- 2018
- Full Text
- View/download PDF
22. Incidence of Comorbidities in Women with Breast Cancer Treated with Tamoxifen or an Aromatase Inhibitor: An Australian Population-Based Cohort Study
- Author
-
David Roder, Theo Niyonsenga, Agnes Vitry, Huah Shin Ng, Bogda Koczwara, Ng, Huah Shin, Koczwara, Bogda, Roder, David, Niyonsenga, Theo, and Vitry, Agnes
- Subjects
Oncology ,medicine.medical_specialty ,medicine.drug_class ,lcsh:Medicine ,comorbidities ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,Epidemiology of cancer ,Epidemiology ,medicine ,030212 general & internal medicine ,hormone-dependent breast cancer ,Aromatase inhibitor ,tamoxifen ,endocrine therapy ,business.industry ,Incidence (epidemiology) ,lcsh:R ,Cancer ,medicine.disease ,030220 oncology & carcinogenesis ,aromatase inhibitor ,Original Article ,business ,Tamoxifen ,cancer epidemiology ,Cohort study ,medicine.drug - Abstract
Background The development of comorbidities has become increasingly relevant with longer-term cancer survival. Objective To assess the pattern of comorbidities among Australian women with breast cancer treated with tamoxifen or an aromatase inhibitor. Design Retrospective cohort study using Pharmaceutical Benefits Scheme (PBS) data (10% sample) from January 2003 to December 2014. Dispensing claims data were used to identify comorbidities and classified with the Rx-Risk-V model. The breast cancer cohort had tamoxifen or an aromatase inhibitor dispensed between 2004 and 2011 with no switching between types of endocrine therapy. Comparisons were made between the breast cancer cohort and specific control groups (age- and sex-matched at 1:10 ratio without any dispensing of anti-neoplastic agents during the study period) for the development of five individual comorbidities over time using Cox regression models. Results Women treated with tamoxifen had a higher incidence of cardiovascular conditions, diabetes, and pain or pain-inflammation, but a lower incidence of hyperlipidaemia compared with non-cancer control groups, as indicated by PBS data. Women treated with aromatase inhibitors were more likely to develop cardiovascular conditions, osteoporosis, and pain or pain-inflammation compared with non-cancer control groups. The risks of hyperlipidaemia and osteoporosis were significantly lower among tamoxifen users compared with aromatase inhibitor users. Conclusions Women with hormone-dependent breast cancer treated with an endocrine therapy had a higher risk of developing specified comorbid conditions than women without cancer, with different comorbidity profiles for those on tamoxifen versus aromatase inhibitors. Further research into the causes and mechanism of development and management of comorbidities after cancer is needed.
- Published
- 2018
- Full Text
- View/download PDF
23. Comorbidities in Australian women with hormone‐dependent breast cancer: a population‐based analysis
- Author
-
Huah Shin Ng, Theo Niyonsenga, David Roder, Bogda Koczwara, Agnes Vitry, Ng, Huah Shin, Koczwara, Bogda, Roder, David M, Niyonsenga, Theo, and Vitry, Agnes I
- Subjects
medicine.medical_specialty ,Antineoplastic Agents, Hormonal ,Osteoporosis ,Breast Neoplasms ,Comorbidity ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Diabetes mellitus ,Internal medicine ,medicine ,Humans ,030212 general & internal medicine ,hormone-dependent breast cancer ,Depression (differential diagnoses) ,Aged ,Retrospective Studies ,Aged, 80 and over ,endocrine therapy ,Depression ,business.industry ,Hazard ratio ,Australia ,Cancer ,General Medicine ,Middle Aged ,medicine.disease ,Cardiovascular Diseases ,030220 oncology & carcinogenesis ,Chronic Disease ,Female ,business ,Cohort study - Abstract
Objective: To compare how frequently selected chronic diseases developed in women with breast cancer receiving endocrine therapy, and in women without cancer. Design, setting and participants: Retrospective, rolling cohort study, analysing a random 10% sample of Pharmaceutical Benefits Scheme (PBS) data for the period 1 January 2003 – 31 December 2014. Women with breast cancer who first commenced endocrine therapy between January 2004 and December 2011 were identified, and age- and sex-matched (1:10) by comorbidity with control groups of women who did not have a dispensing record for antineoplastic agents during the study period or the comorbidity of interest at baseline. Main outcome measures: Development of any of eight pre-selected comorbidities, identified in PBS claims data with the RxRisk-V model. Results: Women with hormone-dependent breast cancer were significantly more likely than women in the control group to develop depression (overall hazard ratio [HR], 1.36; 95% CI, 1.26–1.46), pain or pain–inflammation (HR, 1.30; 95% CI, 1.23–1.38), osteoporosis (overall HR, 1.27; 95% CI, 1.17–1.39), diabetes (HR, 1.24; 95% CI, 1.10–1.41), cardiovascular disorders (overall HR, 1.22; 95% CI, 1.13–1.32), and gastric acid disorders (HR, 1.20; 95% CI, 1.13–1.28). The hazard ratios for developing cardiovascular disorders, depression and osteoporosis were highest during the first year of endocrine therapy. The risk of hyperlipidaemia was lower among women with breast cancer than in the control group (HR, 0.88; 95% CI, 0.81–0.96). There was no significant difference between the two groups in the risk of reactive airway diseases (HR, 1.05; 95% CI, 0.98–1.13). Conclusion: Comorbid conditions are more likely to develop in women who have been diagnosed with hormone-dependent breast cancer than in women without cancer. Our results further support the need to develop appropriate models of care to manage the multiple chronic disorders of breast cancer survivors. Refereed/Peer-reviewed
- Published
- 2018
- Full Text
- View/download PDF
24. Estrogenic and antiestrogenic activities of 2,4-diphenylfuran-based ligands of estrogen receptors α and β
- Author
-
Zimmermann, Jochen and von Angerer, Erwin
- Subjects
- *
STEROID hormones , *ESTROGEN antagonists , *ESTROGEN receptors , *SEX hormones - Abstract
Abstract: The estrogen receptor (ER) exists in two isoforms ERα and ERβ with a different distribution in the body and different functions which are not clearly identified yet. Thus, it is desirable to have both agonists and antagonists with selectivity for one or the other ER isoform available. In a previous study we showed that 2,5-diphenylfurans can be converted into pure antiestrogens with preference for ERα. When the arrangement of the phenyl rings was altered to a 2,4-substitution, the α-selectivity was lost as demonstrated by comparative assays using recombinant human ERα and ERβ. 3,5-Dialkyl-2,4-bis(4-hydroxyphenylfurans) were shown to act as agonists with preference for ERβ. Replacement of one of the alkyl groups by the [(pentylsulfanyl)propyl]aminohexyl side chain afforded estrogen antagonists without receptor selectivity. These derivatives were characterized as pure antiestrogens in transcription and proliferation assays in ER+ MCF-7 breast cancer cells. The most potent antagonists displayed IC50 values of ca. 20nM (fulvestrant 4nM). The data showed that the 2,4-arrangement of the phenyl rings in the furan structure increases the binding affinity for ERβ in comparison to the isomeric 2,5-diphenylfurans but does not lead to a pure antagonist with selectivity for ERβ. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
25. 2,5-Diphenylfuran-based pure antiestrogens with selectivity for the estrogen receptor α
- Author
-
Zimmermann, Jochen, Liebl, Renate, and von Angerer, Erwin
- Subjects
- *
ESTROGEN receptors , *SEX hormones , *BREAST cancer , *CANCER cells , *STEROLS - Abstract
Abstract: The estrogen receptor α (ERα) is understood to play an important role in the progression of breast cancer. Therefore, pure antiestrogens with a preference for this receptor form are of interest as new agents for the treatment of this malignancy. Several chemical structures with selective binding affinity for ERα have been identified and might be useful for the synthesis of ERα-selective pure antiestrogens. In this study we applied the 2,5-diphenylfuran system which is closely related to the triphenylfurans described by others. Various side chains with amino and/or sulfur functions were linked to C3 to convert the furans to estrogen antagonists without residual estrogenic activity. The degree of α-selectivity which ranges from 2.5- to 236-fold is strongly influenced by the alkyl group at C4. Antiestrogenic potency was determined in MCF-7/2a breast cancer cells stably transfected with a luciferase gene under the control of an ERE. The 2,5-bis(4-hydroxyphenyl)furan with an ethyl substituent and a 6-[N-methyl-N-(3-pentylthiopropyl)amino]hexyl side chain exerted the strongest antiestrogenic effect in this series with an IC50 value of 50nM in cells stimulated with 1nM estradiol. The RBA values of this derivative were 18% (ERα) and 3.4% (ERβ) of estradiol, respectively. It inhibited the growth of wild-type MCF-7 cells with an IC50 value of 22nM. The data show that the 2,5-diphenylfuran system is appropriate for the development of pure antiestrogens with preference for ERα. [Copyright &y& Elsevier]
- Published
- 2005
- Full Text
- View/download PDF
26. Nuclear PDCD4 Expression Defines a Subset of Luminal B-Like Breast Cancers with Good Prognosis
- Author
-
Cecilia J. Proietti, Sandra L. Ares, Ernesto Gil Deza, Pablo Guzmán, Juan Carlos Roa, Agustina Dupont, Agustina Roldán Deamicis, Roxana Schillaci, Rosalia Ines Cordo Russo, Sabrina Barchuk, Franco Izzo, Felipe G. Gercovich, Matías G. Pereyra, Leandro Venturutti, Fabiana Anfuso, Patricia V. Elizalde, Eduardo Cortese, Teresa Castiglioni, Violeta A. Chiauzzi, Mauro E. Cenciarini, Santiago Madera, Gabriel Lebersztein, Silvina Figurelli, Claudio Levit, María F. Chervo, and Daniel Lopez Della Vecchia
- Subjects
0301 basic medicine ,Cancer Research ,medicine.drug_class ,Otras Ciencias Biológicas ,Endocrinology, Diabetes and Metabolism ,BIOMARKERS ,Luma ,Breast Neoplasms ,Disease ,HORMONE-DEPENDENT BREAST CANCER ,Ciencias Biológicas ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Breast cancer ,BREAST CANCER ,Progesterone receptor ,Medicine ,Gene silencing ,Humans ,LUMINAL B-LIKE BREAST CANCER ,PDCD4 ,Endocrine and Autonomic Systems ,business.industry ,RNA-Binding Proteins ,medicine.disease ,Prognosis ,030104 developmental biology ,Oncology ,Estrogen ,030220 oncology & carcinogenesis ,Cancer research ,Biomarker (medicine) ,Immunohistochemistry ,Female ,TUMOR SUPPRESSOR ,business ,Apoptosis Regulatory Proteins ,CIENCIAS NATURALES Y EXACTAS - Abstract
The hormone receptor-positive (estrogen and/or progesterone receptor (PR)-positive) and HER2-negative breast cancer (BC) subtype is a biologically heterogeneous entity that includes luminal A-like (LumA-like) and luminal B-like (LumB-like) subtypes. Decreased PR levels is a distinctive biological feature of LumB-like tumors. These tumors also show reduced sensitivity to endocrine therapies and poorer prognosis than LumA-like tumors. Identification of biomarkers to accurately predict disease relapse in these subtypes is crucial in order to select effective therapies. We identified the tumor suppressor PDCD4 (programmed cell death 4), located in the nucleus (NPDCD4), as an independent prognostic factor of good clinical outcome in LumA-like and LumB-like subtypes. NPDCD4-positive LumB-like tumors presented overall and disease-free survival rates comparable to those of NPDCD4-positive LumA-like tumors, indicating that NPDCD4 improves the outcome of LumB-like patients. In contrast, NPDCD4 loss increased the risk of disease recurrence and death in LumB-like compared with LumA-like tumors. This, along with our results showing that LumB-like tumors present lower NPDCD4 positivity than LumA-like tumors, suggests that NPDCD4 loss contributes to endocrine therapy resistance in LumB-like BCs. We also revealed that PR induces PDCD4 transcription in LumB-like BC, providing a mechanistic explanation to the low PDCD4 levels in LumB-like BCs lacking PR. Finally, PDCD4 silencing enhanced BC cell survival in a patient-derived explant model of LumB-like disease. Our discoveries highlight NPDCD4 as a novel biomarker in LumA- and LumB-like subtypes, which could be included in the panel of immunohistochemical markers used in the clinic to accurately predict the prognosis of LumB-like tumors. Fil: Madera, Santiago. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina Fil: Chervo, María Florencia. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina Fil: Chiauzzi, Violeta Alicia. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina Fil: Pereyra Matías G.. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina Fil: Venturutti, Leandro. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina Fil: Izzo, Franco. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina Fil: Roldán Deamicis, Agustina. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina Fil: Guzmán, Pablo. Universidad de La Frontera. Núcleo Científico y Tecnológico en Recursos Naturales; Chile Fil: Dupont, Agustina. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina Fil: Roa, Juan Carlos. Universidad de La Frontera. Núcleo Científico y Tecnológico en Recursos Naturales; Chile. Universidad Católica de Chile; Chile Fil: Cenciarini, Mauro Ezequiel. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina Fil: Barchuk, Sabrina. Gobierno de la Ciudad de Buenos Aires. Hospital General de Agudos "Juan A. Fernández"; Argentina Fil: Figurelli, Silvina. Gobierno de la Ciudad de Buenos Aires. Hospital General de Agudos "Juan A. Fernández"; Argentina Fil: Lopez Della Vecchia, Daniel Edgardo. Gobierno de la Ciudad de Buenos Aires. Hospital General de Agudos "Juan A. Fernández"; Argentina Fil: Levit, Claudio. Sanatorio Sagrado Corazón. Servicio de Ginecología; Argentina Fil: Lebersztein, Gabriel. Sanatorio Sagrado Corazón. Servicio de Ginecología; Argentina Fil: Anfuso, Fabiana. Sanatorio Sagrado Corazón. Servicio de Ginecología; Argentina Fil: Castiglioni, Teresa. Centro de Patología Dr. Elsner; Argentina Fil: Cortese, Eduardo Mateo. Hospital Aeronáutico Central. Servicio de Ginecología; Argentina Fil: Ares, Sandra. Instituto Oncológico Henry Moore; Argentina Fil: Gil Deza, Ernesto. Instituto Oncológico Henry Moore; Argentina Fil: Gercovich, Felipe G.. Instituto Oncológico Henry Moore; Argentina Fil: Proietti Anastasi, Cecilia Jazmín. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina Fil: Schillaci, Roxana. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina Fil: Cordo Russo, Rosalía I.. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina Fil: Elizalde, Patricia Virginia. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina
- Published
- 2020
27. Novel D-ring modified steroid derivatives as potent, non-estrogenic, steroid sulfatase inhibitors with in vivo activity
- Author
-
Fischer, Delphine S., Chander, Surinder K., Woo, L.W. Lawrence, Fenton, Janine C., Purohit, Atul, Reed, Michael J., and Potter, Barry V.L.
- Subjects
- *
STEROIDS , *BREAST cancer - Abstract
In pursuit of novel steroid sulfatase (STS) inhibitors devoid of estrogenicity, several D-ring modified steroid derivatives were synthesised. In vitro evaluation of the compounds identified two highly potent inhibitors, 4a and 4b, which were 18 times more active than estrone-3-O-sulfamate (EMATE), both having IC50 values of ca. 1 nM. These 16,17-seco-estra-1,3,5(10)-triene-16,17-imide derivatives were synthesised from estrone, via the intermediate 1, which was easily alkylated, deprotected and sulfamoylated affording the final compounds in high yields. In order to assess their biological profile, the selected inhibitors were tested for their in vivo inhibitory potency and estrogenicity in ovariectomised rats. After an oral dose of 10 mg/kg per day for 5 days, 4a and 4b were found to inhibit rat liver steroid sulfatase by 99%. They were also devoid of estrogenic activity in the uterine weight gain assay, indicating that these two leads have therapeutic potential for the treatment of hormone-dependent breast cancer. [Copyright &y& Elsevier]
- Published
- 2003
- Full Text
- View/download PDF
28. Pharmacocinétique et pharmacogénétique du tamoxifène et du létrozole chez les patientes atteintes d’un cancer du sein hormonodépendant
- Author
-
Puszkiel, Alicja, Centre de Recherches en Cancérologie de Toulouse (CRCT), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Université Paul Sabatier - Toulouse III, Mélanie White-Koning, and Fabienne Thomas
- Subjects
Hormone-Dependent breast cancer ,Tamoxifen ,Pharmacogenetics ,Pharmacocinétique ,Letrozole ,Tamoxifène ,[SDV.SP.PHARMA]Life Sciences [q-bio]/Pharmaceutical sciences/Pharmacology ,Pharmacogénétique ,Pharmacokinetics ,Cancer du sein hormonodépendant ,Létrozole - Abstract
Tamoxifen and aromatase inhibitors are the main therapeutic options for adjuvant treatment of hormone-dependent breast cancer. High inter-individual variability of efficacy and toxicity of these drugs is observed which may be related to the variability of plasma drug concentrations. In the case of tamoxifen, its pharmacological activity depends on the formation of active metabolites, mainly endoxifen, via the cytochromes P450 (CYP2D6, CYP3A4/5, CYP2C9, CYP2C19). Concerning letrozole, it is metabolised via the CYP2A6 and CYP3A4 isoenzymes. The variable activity of CYP, due to genetic polymorphisms or concomitant use of CYP inhibitors or inducers, may impact the plasma exposure to the active compounds and in fine, the efficacy and toxicity of the treatment. The pharmacokinetic-pharmacogenetic analyses of the data from first follow-up visit in the PHACS study (prospective, multicenter, national, 2000 patients, 6 follow-up visits during 3 years) demonstrated the impact of CYP2D6 phenotype, CYP3A4*22, CYP2C19*2, and CYP2B6*6 genotypes and of the concomitant use of CYP2D6 inhibitors on tamoxifen metabolism. The longitudinal pharmacokinetic data for tamoxifen and its metabolites (6 to 36 months follow-up visits) were analysed simultaneously using non-linear mixed effects modelling (population approach). A joint pharmacokinetic model for tamoxifen and six metabolites was developed and validated. The significant covariates in the final model were: CYP2D6 phenotype, CYP3A4*22 and CYP2B6*6 genotypes, CYP2D6 inhibitors, age and body weight. Simulations of alternative dosing regimens were performed to propose an individualised dose of tamoxifen for patients at risk of subtherapeutic exposure to endoxifen. The pharmacokinetic-pharmacogenetic data for letrozole were analysed using the population approach. A one-compartment model was developed and validated. CYP2A6 activity was significantly associated with letrozole clearance. The model was used for identification of non-adherent patients based on their steady-state plasma concentrations.; Le tamoxifène et les inhibiteurs de l’aromatase constituent les principaux médicaments en hormonothérapie utilisées dans le traitement adjuvant des cancers du sein hormono-dépendants. Une grande variabilité de l’efficacité et de la toxicité de ces traitements est observée et pourrait être liée à des différences de concentration du médicament au niveau du plasma. Dans le cas du tamoxifène, son activité pharmacologique est dépendante de la formation de métabolites actifs, notamment de l’endoxifène, par l’intermédiaire des cytochromes de la famille P450 (CYP2D6, CYP3A4/5, CYP2C9, CYP2C19). Le létrozole, quant à lui, est métabolisé par le CYP2A6 et le CYP3A4. La variabilité de l’activité enzymatique de ces enzymes, due notamment aux polymorphismes génétiques et à la prise concomitante d’inhibiteurs ou d’inducteurs enzymatiques, pourrait avoir un impact sur les concentrations plasmatiques et in fine, sur l’efficacité et la toxicité du traitement. Les analyses pharmacocinétiques-pharmacogénétiques sur les données de la première visite de suivi de l’étude PHACS (étude prospective, multicentrique, nationale, 2000 patientes, 6 visites de suivi pendant 3 ans) ont démontré l’impact du phénotype CYP2D6, génotypes CYP3A4*22, CYP2C19*2 et CYP2B6*6 ainsi que de la prise concomitante des inhibiteurs de CYP2D6 sur la métabolisation du tamoxifène. L’analyse simultanée des concentrations plasmatiques du tamoxifène et de ses métabolites sur une durée de 3 ans (visites de 6 à 36 mois) a été effectuée par des modèles non-linéaires à effets mixtes (approche de pharmacocinétique de population). Un modèle pharmacocinétique décrivant les concentrations de tamoxifène et de six métabolites a été développé et validé. Les covariables significatives dans le modèle final étaient: le phénotype CYP2D6, les génotypes CYP3A4*22 et CYP2B6*6, les inhibiteurs de CYP2D6, l’âge et le poids. Des simulations de schémas d’administration alternatifs ont été effectuées afin de proposer des doses adaptées aux patientes à risque de sous-exposition à l’endoxifène. Les données pharmacocinétiques et pharmacogénétiques du létrozole ont été analysées par approche de pharmacocinétique de population. Un modèle monocompartimental a été développé et validé. L’activité de CYP2A6 était significativement associée à la clairance du létrozole. Le modèle pharmacocinétique a été utilisé pour identifier les patientes non-adhérentes au traitement en se basant sur leurs concentrations plasmatiques à l’état d’équilibre.
- Published
- 2019
29. Pharmacokinetics and pharmacogenetics of tamoxifen and letrozole in patients with hormone-dependent breast cancer
- Author
-
Puszkiel, Alicja, Centre de Recherches en Cancérologie de Toulouse (CRCT), Centre National de la Recherche Scientifique (CNRS)-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paul Sabatier - Toulouse III, Mélanie White-Koning, Fabienne Thomas, Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and STAR, ABES
- Subjects
Hormone-Dependent breast cancer ,Tamoxifen ,Pharmacogenetics ,Pharmacocinétique ,[SDV.SP.PHARMA] Life Sciences [q-bio]/Pharmaceutical sciences/Pharmacology ,Letrozole ,Tamoxifène ,[SDV.SP.PHARMA]Life Sciences [q-bio]/Pharmaceutical sciences/Pharmacology ,Pharmacogénétique ,Pharmacokinetics ,Cancer du sein hormonodépendant ,Létrozole - Abstract
Tamoxifen and aromatase inhibitors are the main therapeutic options for adjuvant treatment of hormone-dependent breast cancer. High inter-individual variability of efficacy and toxicity of these drugs is observed which may be related to the variability of plasma drug concentrations. In the case of tamoxifen, its pharmacological activity depends on the formation of active metabolites, mainly endoxifen, via the cytochromes P450 (CYP2D6, CYP3A4/5, CYP2C9, CYP2C19). Concerning letrozole, it is metabolised via the CYP2A6 and CYP3A4 isoenzymes. The variable activity of CYP, due to genetic polymorphisms or concomitant use of CYP inhibitors or inducers, may impact the plasma exposure to the active compounds and in fine, the efficacy and toxicity of the treatment. The pharmacokinetic-pharmacogenetic analyses of the data from first follow-up visit in the PHACS study (prospective, multicenter, national, 2000 patients, 6 follow-up visits during 3 years) demonstrated the impact of CYP2D6 phenotype, CYP3A4*22, CYP2C19*2, and CYP2B6*6 genotypes and of the concomitant use of CYP2D6 inhibitors on tamoxifen metabolism. The longitudinal pharmacokinetic data for tamoxifen and its metabolites (6 to 36 months follow-up visits) were analysed simultaneously using non-linear mixed effects modelling (population approach). A joint pharmacokinetic model for tamoxifen and six metabolites was developed and validated. The significant covariates in the final model were: CYP2D6 phenotype, CYP3A4*22 and CYP2B6*6 genotypes, CYP2D6 inhibitors, age and body weight. Simulations of alternative dosing regimens were performed to propose an individualised dose of tamoxifen for patients at risk of subtherapeutic exposure to endoxifen. The pharmacokinetic-pharmacogenetic data for letrozole were analysed using the population approach. A one-compartment model was developed and validated. CYP2A6 activity was significantly associated with letrozole clearance. The model was used for identification of non-adherent patients based on their steady-state plasma concentrations., Le tamoxifène et les inhibiteurs de l’aromatase constituent les principaux médicaments en hormonothérapie utilisées dans le traitement adjuvant des cancers du sein hormono-dépendants. Une grande variabilité de l’efficacité et de la toxicité de ces traitements est observée et pourrait être liée à des différences de concentration du médicament au niveau du plasma. Dans le cas du tamoxifène, son activité pharmacologique est dépendante de la formation de métabolites actifs, notamment de l’endoxifène, par l’intermédiaire des cytochromes de la famille P450 (CYP2D6, CYP3A4/5, CYP2C9, CYP2C19). Le létrozole, quant à lui, est métabolisé par le CYP2A6 et le CYP3A4. La variabilité de l’activité enzymatique de ces enzymes, due notamment aux polymorphismes génétiques et à la prise concomitante d’inhibiteurs ou d’inducteurs enzymatiques, pourrait avoir un impact sur les concentrations plasmatiques et in fine, sur l’efficacité et la toxicité du traitement. Les analyses pharmacocinétiques-pharmacogénétiques sur les données de la première visite de suivi de l’étude PHACS (étude prospective, multicentrique, nationale, 2000 patientes, 6 visites de suivi pendant 3 ans) ont démontré l’impact du phénotype CYP2D6, génotypes CYP3A4*22, CYP2C19*2 et CYP2B6*6 ainsi que de la prise concomitante des inhibiteurs de CYP2D6 sur la métabolisation du tamoxifène. L’analyse simultanée des concentrations plasmatiques du tamoxifène et de ses métabolites sur une durée de 3 ans (visites de 6 à 36 mois) a été effectuée par des modèles non-linéaires à effets mixtes (approche de pharmacocinétique de population). Un modèle pharmacocinétique décrivant les concentrations de tamoxifène et de six métabolites a été développé et validé. Les covariables significatives dans le modèle final étaient: le phénotype CYP2D6, les génotypes CYP3A4*22 et CYP2B6*6, les inhibiteurs de CYP2D6, l’âge et le poids. Des simulations de schémas d’administration alternatifs ont été effectuées afin de proposer des doses adaptées aux patientes à risque de sous-exposition à l’endoxifène. Les données pharmacocinétiques et pharmacogénétiques du létrozole ont été analysées par approche de pharmacocinétique de population. Un modèle monocompartimental a été développé et validé. L’activité de CYP2A6 était significativement associée à la clairance du létrozole. Le modèle pharmacocinétique a été utilisé pour identifier les patientes non-adhérentes au traitement en se basant sur leurs concentrations plasmatiques à l’état d’équilibre.
- Published
- 2019
30. L'influence des insecticides néonicotinoïdes sur la stéroïdogenèse dans deux modèles cellulaires humains en co-culture physiologiquement représentatifs
- Author
-
Viau, Rachel and Viau, Rachel
- Abstract
Il existe des preuves croissantes qui démontrent que les humains sont constamment exposés à des composés chimiques perturbateurs endocriniens via l’alimentation et l’environnement. Une nouvelle classe majeure d’insecticides utilisés dans le monde pour la protection des cultures agricoles, nommée les néonicotinoïdes, est de plus en plus présent dans l’environnement. Le thiamethoxame, le thiaclopride et l’imidaclopride sont les plus couramment utilisés sur le marché. Leurs persistances et applications posent plusieurs inquiétudes pour la santé humaine. Les perturbateurs endocriniens sont connus pour affecter les processus hormonaux qui pourraient causer des problèmes à la reproduction, des toxicités ainsi que de promouvoir certains cancers, comme le cancer du sein hormono-dépendant. L’aromatase (CYP19) est une enzyme clé de la synthèse des oestrogènes, où le gène est exprimé de façon promoteur- et tissu-spécifique. Cette enzyme peut être problématique dans le développement du cancer du sein hormono-dépendant puisque la progression est basée sur la surproduction d’oestrogènes des cellules stromales aux alentours des tumeurs. L’aromatase est aussi essentielle au bon développement foetal lors de la grossesse en assurant les communications complexes entre la mère, le placenta et le foetus. L’objectif de cette étude était de démystifier les mécanismes que les insecticides néonicotinoïdes utilisent pour causer leurs effets potentiels perturbateurs endocriniens dans deux modèles de co-cultures distinctes et physiologiquement représentatives. En premier lieu, nous avons clarifiés les mécanismes par lesquels ces contaminants perturbent la biosynthèse de l’oestriol dans un modèle de co-culture féto-placentaire utilisant les lignées cellulaires BeWo (placentaires) et H295R (surrénale foetale). Notre deuxième objectif était de développer un nouveau modèle de co-culture représentant le microenvironnement stroma-tumoral d’un cancer du sein oestrogène-dépendant en utilisant les lign
- Published
- 2018
31. Novel Selective Estrogen Receptor Ligand Conjugates Incorporating Endoxifen-Combretastatin and Cyclofenil-Combretastatin Hybrid Scaffolds: Synthesis and Biochemical Evaluation
- Author
-
Mary J. Meegan, Bassem Yassin, Darren Fayne, Daniela M. Zisterer, Gloria Ana, Niall O. Keely, Patrick M. Kelly, and Sandra A. Bright
- Subjects
0301 basic medicine ,Models, Molecular ,Molecular Conformation ,Pharmaceutical Science ,Estrogen receptor ,Pharmacology ,Crystallography, X-Ray ,Ligands ,Analytical Chemistry ,chemistry.chemical_compound ,0302 clinical medicine ,Drug Discovery ,Bibenzyls ,Cytotoxicity ,hormone-dependent breast cancer ,apoptosis ,tumour targeting conjugates ,selective estrogen receptor modulators ,combretastatin A-4(CA-4) ,endoxifen ,cyclofenil ,estrogen receptor ligands ,Ligand (biochemistry) ,G2 Phase Cell Cycle Checkpoints ,Receptors, Estrogen ,Chemistry (miscellaneous) ,Selective estrogen receptor modulator ,030220 oncology & carcinogenesis ,MCF-7 Cells ,Molecular Medicine ,Protein Binding ,Article ,lcsh:QD241-441 ,03 medical and health sciences ,Inhibitory Concentration 50 ,lcsh:Organic chemistry ,Cyclofenil ,Humans ,Physical and Theoretical Chemistry ,Cell Proliferation ,Combretastatin ,Organic Chemistry ,Antineoplastic Agents, Phytogenic ,Tamoxifen ,030104 developmental biology ,chemistry ,Nuclear receptor ,Cancer research ,Leukocytes, Mononuclear ,Drug Screening Assays, Antitumor ,Conjugate - Abstract
Nuclear receptors such as the estrogen receptors (ERα and ERβ) modulate the effects of the estrogen hormones and are important targets for design of innovative chemotherapeutic agents for diseases such as breast cancer and osteoporosis. Conjugate and bifunctional compounds which incorporate an ER ligand offer a useful method of delivering cytotoxic drugs to tissue sites such as breast cancers which express ERs. A series of novel conjugate molecules incorporating both the ER ligands endoxifen and cyclofenil-endoxifen hybrids covalently linked to the antimitotic and tubulin targeting agent combretastatin A-4 were synthesised and evaluated as ER ligands. A number of these compounds demonstrated pro-apoptotic effects, with potent antiproliferative activity in ER-positive MCF-7 breast cancer cell lines and low cytotoxicity. These conjugates displayed binding affinity towards ERα and ERβ isoforms at nanomolar concentrations e.g., the cyclofenil-amide compound 13e is a promising lead compound of a clinically relevant ER conjugate with IC50 in MCF-7 cells of 187 nM, and binding affinity to ERα (IC50 = 19 nM) and ERβ (IC50 = 229 nM) while the endoxifen conjugate 16b demonstrates antiproliferative activity in MCF-7 cells (IC50 = 5.7 nM) and binding affinity to ERα (IC50 = 15 nM) and ERβ (IC50 = 115 nM). The ER binding effects are rationalised in a molecular modelling study in which the disruption of the ER helix-12 in the presence of compounds 11e, 13e and 16b is presented These conjugate compounds have potential application for further development as antineoplastic agents in the treatment of ER positive breast cancers.
- Published
- 2017
32. President’s address: Treatment of tamoxifen-refractory breast cancer — approach by animal models.
- Author
-
Iino, Yuichi
- Published
- 2001
- Full Text
- View/download PDF
33. Dual Aromatase-Sulphatase Inhibitors (DASIs) for the Treatment of Hormone Dependent Breast Cancer.
- Author
-
Banjare L, Jain AK, and Thareja S
- Subjects
- Aromatase metabolism, Estrogens, Female, Humans, Sulfatases metabolism, Aromatase Inhibitors pharmacology, Aromatase Inhibitors therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms metabolism
- Abstract
Breast cancer is the most frequently diagnosed cancer in women and the second most common form of cancer, causing death after lung cancer, all across the globe at an alarming rate. The level of estrogens in breast cancer tissues of postmenopausal women is 10-40 folds higher than the non-carcinogenic breast tissues. As a result of this greater level of estrogen, breast tissue becomes more prone to develop breast cancer; mainly, estradiol plays a significant role in the initiation and development of hormone-dependent breast cancer. Androstenedione, Adrenal dehydroepiandrosterone sulfate, and estrone-sulfate also play an important role as precursors for estrogen biosynthesis. Estrogen deprivation exhibits an attractive phenomenon in the advancement of ideal therapeutics for the treatment of breast cancer. Inhibition of aromatase and sulphatase emerged as an attractive therapy for the treatment of hormone-dependent breast cancer via deprivation of estrogen by different pathways. The cocktail of aromatase and sulphatase inhibitors known as Dual Aromatase-sulphatase Inhibitors (DASIs) emerged as an attractive approach for effective estrogen deprivation. The present review article focused on the journey of dual aromatase-sulphatase inhibitors from the beginning to date (2020). Keeping in view the key observations, this review may be helpful for medicinal chemists to design and develop new and efficient dual aromatase-sulphatase inhibitors for the possible treatment of hormone- related breast cancer., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.net.)
- Published
- 2021
- Full Text
- View/download PDF
34. Novel 2-substituted isoflavones: A privileged structure approach to new agents for hormone-dependent breast cancer
- Author
-
Kim, Young-Woo
- Subjects
- hormone-dependent breast cancer, estrogens, isoflavones, aromatase inhibitors, selective estrogen receptor modulators, 2-substituted isoflavones, privileged structure
- Abstract
Natural isoflavones are well known as phytoestrogens due to their biological activities that mimic endogenous estrogens. For this reason, we recognized the isoflavone nucleus as a promising privileged structure for the development of new therapeutics for hormone-dependent breast cancer, in which estrogens play a key role in growth and development of tumor. We envisioned that a specific activity could be achieved by introducing proper functional groups into the isoflavone nucleus, and we designed a library of novel 2-substituted isoflavones. For the library synthesis, we developed efficient synthetic routes, in which alpha-oxoketene dithioacetals are employed as key intermediates. Various 2-(alkylthio)isoflavones were obtained directly from readily available deoxybenzoins and electrophiles using a phase transfer catalysis procedure. Alternatively, 2-substituted isoflavones were prepared through a 1,4-conjugate addition-elimination reaction of 2-(methylsulfonyl)isoflavones using a variety of commercially available nucleophiles. With efficient synthetic routes developed, we examined the potential utility of this approach for the discovery of new leads towards specific molecular targets in the breast cancer. Initially, we have focused on two major classes of therapeutic agents, aromatase inhibitors and selective estrogen receptor modulators (SERMs). As potential aromatase inhibitors, we prepared various isoflavones possessing a nitrogen-containing heterocycle, which may interfere with aromatase activity by coordinating with its heme iron. Several compounds were identified with potent aromatase inhibitory activities. As potential SERMs, we prepared a series of isoflavones containing an amine-bearing side chain, which is known to be essential for the tissue selectivity of many known SERMs. Several compounds in this series were highly potent in inhibiting proliferation of human breast cancer cells. However, their low binding affinities for estrogen receptors suggest that these compounds may not be SERMs. Currently, extended studies are underway to elucidate their mechanisms of action. Consequently, our approach using isoflavone as a privileged structure proved useful to identify new leads for breast cancer treatment. In addition, with a growing number of newly identified proteins in this post-genome era, the number of potential targets for therapeutic intervention has also increased. Therefore, our approach could also be useful in identifying new chemical probes that can provide insight into biological problems presented by breast cancer.
- Published
- 2003
35. Stimulation of breast cancer cellsin vitro by the environmental estrogen enterolactone and the phytoestrogen equol
- Author
-
Welshons, W. V., Murphy, C. S., Koch, R., Calaf, G., and Jordan, V. C.
- Published
- 1987
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.