Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
1.
Steroids ; 191: 109163, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36581086

RESUMEN

Curcumin is a phytochemical derived from the spice turmeric that is reported to have therapeutic effects. We are studying the enzyme steroid sulfatase (STS), which removes the sulfate group from inactive steroid hormones in peripheral tissues and we were interested in the effect of curcumin on STS activity due to its structural composition (polyphenolic). We sought to determine if curcumin affects STS activity in two model systems, rat liver and NIH-3T3 mouse fibroblast cells. STS assays were performed on tissue extracts of rat liver, and on NIH-3T3 microsomes and cells, with and without curcumin. Male and female rat liver extracts contained substantial amounts of STS activity, with males averaging higher (4-11 %) levels. Estradiol inhibited STS activity in livers of both sexes at 20 and 10 µM. Curcumin acted as a competitive inhibitor of STS activity in rat liver extracts, with a Ki of 19.8 µM in males and 9.3 µM in females. Curcumin also inhibited STS activity in NIH-3T3 microsomes at both 20 µM and 10 µM, and in whole NIH-3T3 cells at 20 µM. These data are the first to demonstrate STS inhibition by curcumin. Inhibition of STS results in lower active steroid hormone (estrogens and androgens) levels in tissues, possibly altering modulation of immune responses by these steroids.


Asunto(s)
Curcumina , Inhibidores Enzimáticos , Esteril-Sulfatasa , Animales , Femenino , Masculino , Ratones , Ratas , Curcumina/farmacología , Inhibidores Enzimáticos/farmacología , Hígado/efectos de los fármacos , Hígado/enzimología , Extractos Hepáticos , Células 3T3 NIH , Esteril-Sulfatasa/antagonistas & inhibidores
2.
Bioorg Chem ; 129: 106148, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36244324

RESUMEN

Steroid sulfatase inhibitors block the local production of estrogenic steroids and are attractive agents for the treatment of estrogen-dependent cancers. Inspiration of coumarin-based inhibitors, we synthesized thirty-two 5-oxa-1,2,3,4-tetrahydro-2H-chromeno-(3,4-c)pyridin-8-yl sulfamates, focusing on the substitution derivatives on the adjacent phenyl ring and evaluated their abilities to block STS from human placenta and MCF-7 cells. SAR analysis revealed that the incorporation of chlorine at either meta and/or para position of the adjacent phenyl ring of the tricyclic skeleton enhanced STS inhibition. Di-substitutions at the adjacent phenyl ring were superior to mono and tri-substitutions. Further kinetic analysis of these compounds revealed that chloride-bearing compounds, such as 19m, 19v, and 19w, had KI of 0.02 to 0.11 nM and kinact/KI ratios of 8.8-17.5 nM-1min-1, a parameter indicated for the efficiency of irreversible inhibition. We also used the docking model to illustrate the difference in STS inhibitory potency of compounds. Finally, the safety and anti-cancer activity of selected compounds 19m, 19v, and 19w were also studied, showing the results of low cytotoxicity on NHDF cell line and being more potent than irosustat on ZR-75-1 cell, which was a hormone-dependent cancer cell line with high STS expression.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos , Placenta , Esteril-Sulfatasa , Ácidos Sulfónicos , Femenino , Humanos , Embarazo , Inhibidores Enzimáticos/farmacología , Cinética , Esteril-Sulfatasa/antagonistas & inhibidores , Relación Estructura-Actividad , Ácidos Sulfónicos/química , Ácidos Sulfónicos/farmacología , Placenta/enzimología , Células MCF-7
3.
Molecules ; 26(10)2021 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-34064842

RESUMEN

Steroid sulphatase (STS), involved in the hydrolysis of steroid sulphates, plays an important role in the formation of both active oestrogens and androgens. Since these steroids significantly impact the proliferation of both oestrogen- and androgen-dependent cancers, many research groups over the past 30 years have designed and developed STS inhibitors. One of the main contributors to this field has been Prof. Barry Potter, previously at the University of Bath and now at the University of Oxford. Upon Prof. Potter's imminent retirement, this review takes a look back at the work on STS inhibitors and their contribution to our understanding of sulphate biology and as potential therapeutic agents in hormone-dependent disease. A number of potent STS inhibitors have now been developed, one of which, Irosustat (STX64, 667Coumate, BN83495), remains the only one to have completed phase I/II clinical trials against numerous indications (breast, prostate, endometrial). These studies have provided new insights into the origins of androgens and oestrogens in women and men. In addition to the therapeutic role of STS inhibition in breast and prostate cancer, there is now good evidence to suggest they may also provide benefits in patients with colorectal and ovarian cancer, and in treating endometriosis. To explore the potential of STS inhibitors further, a number of second- and third-generation inhibitors have been developed, together with single molecules that possess aromatase-STS inhibitory properties. The further development of potent STS inhibitors will allow their potential therapeutic value to be explored in a variety of hormone-dependent cancers and possibly other non-oncological conditions.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Esteril-Sulfatasa/antagonistas & inhibidores , Animales , Vías Biosintéticas/efectos de los fármacos , Ensayos Clínicos como Asunto , Desarrollo de Medicamentos , Inhibidores Enzimáticos/química , Humanos , Esteril-Sulfatasa/metabolismo
4.
Steroids ; 172: 108856, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33945801

RESUMEN

17ß-Hydroxysteroid dehydrogenase type 1 (17ß-HSD1) and steroid sulfatase (STS) are involved in the synthesis of the most potent estrogen in the human body, estradiol (E2). These enzymes are known to play a pivotal role in the progression of estrogen-dependent diseases, such as breast cancer and endometriosis. Therefore, the inhibition of 17ß-HSD1 and/or STS represents a promising avenue to modulate the growth of estrogen-dependent tumors or lesions. We recently established the key role of a bromoethyl side chain added at the C3-position of a 16ß-carbamoyl-benzyl-E2 nucleus to covalently inhibit 17ß-HSD1. To extend the structure-activity relationship study to the C16ß-position of this new selective irreversible inhibitor (PBRM), we synthesized a series of analog compounds by changing the nature of the C16ß-side chain but keeping the 2-bromoethyl group at position C3. We determined their 17ß-HSD1 inhibitions in T-47D cells (transformation of E1 into E2), but we did not obtain a stronger 17ß-HSD1 inhibitor than PBRM. Compounds 16 and 17 were found to be more likely to bind to the catalytic site and showed a promising but moderate inhibitory activity with estimated IC50 values of 0.5 and 0.7 µM, respectively (about 10 times higher than PBRM). Interestingly, adding one or two sulfamate groups in the D-ring's surroundings did not significantly decrease compounds' potential to inhibit 17ß-HSD1, but clearly improved their potential to inhibit STS. These results open the door to the development of a new family of steroid derivatives with dual (17ß-HSD1 and STS) inhibiting actions.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Neoplasias de la Mama/tratamiento farmacológico , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Esteril-Sulfatasa/antagonistas & inhibidores , Trientina/química , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Proliferación Celular , Femenino , Humanos , Relación Estructura-Actividad , Células Tumorales Cultivadas
5.
Expert Opin Ther Pat ; 31(6): 453-472, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33783295

RESUMEN

Introduction: Steroid sulfatase (STS) enzyme is responsible for transforming the inactive sulfate metabolites of steroid sex hormones into the active free steroids. Both the deficiency and the over-expression of STS are associated with the pathophysiology of certain diseases. This article provides the readership with a comprehensive review about STS enzyme and its recently reported inhibitors.Areas covered: In the present article, we reviewed the structure, location, and substrates of STS enzyme, physiological functions of STS, and disease states related to over-expression or deficiency of STS enzyme. STS inhibitors reported during the last five years (2016-present) have been reviewed as well.Expert opinion: Irosustat is the most successful STS inhibitor drug candidate so far. It is currently under investigation in clinical trials for treatment of estrogen-dependent breast cancer. Non-steroidal sulfamate is the most favorable scaffold for STS inhibitor design. They can be beneficial for the treatment of hormone-dependent cancers and neurodegenerative disorders without significant estrogenic side effects. Moreover, dual-acting molecules (inhibitors of STS + another synergistic mechanism) can be therapeutically efficient.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Esteril-Sulfatasa/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Inhibidores Enzimáticos/administración & dosificación , Femenino , Humanos , Patentes como Asunto , Esteril-Sulfatasa/metabolismo , Ácidos Sulfónicos/administración & dosificación , Ácidos Sulfónicos/farmacología
6.
J Enzyme Inhib Med Chem ; 36(1): 238-247, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33322953

RESUMEN

In the present work, we report a new class of potent steroid sulphatase (STS) inhibitors based on 6-(1-phenyl-1H-1,2,3-triazol-4-yl)naphthalen-2-yl sulphamate derivatives. Within the set of new STS inhibitors, 6-(1-(1,2,3-trifluorophenyl)-1H-1,2,3-triazol-4-yl)naphthalen-2-yl sulphamate 3L demonstrated the highest activity in the enzymatic assay inhibiting the STS activity to 7.98% at 0.5 µM concentration. Furthermore, to verify whether the obtained STS inhibitors are able to pass through the cellular membrane effectively, cell line experiments have been carried out. We found that the lowest STS activities were measured in the presence of compound 3L (remaining STS activity of 5.22%, 27.48% and 99.0% at 100, 10 and 1 nM concentrations, respectively). The measured STS activities for Irosustat (used as a reference) were 5.72%, 12.93% and 16.83% in the same concentration range. Moreover, a determined IC50 value of 15.97 nM for 3L showed that this compound is a very promising candidate for further preclinical investigations.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Esteril-Sulfatasa/antagonistas & inhibidores , Ácidos Sulfónicos/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Células MCF-7 , Estructura Molecular , Esteril-Sulfatasa/aislamiento & purificación , Esteril-Sulfatasa/metabolismo , Relación Estructura-Actividad , Ácidos Sulfónicos/síntesis química , Ácidos Sulfónicos/química
7.
J Recept Signal Transduct Res ; 41(2): 123-137, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32705921

RESUMEN

Aromatase and steroidal sulfatase (STS) are steroidogenic enzyme that increases the concentration of estrogens in circulation, a primary factor leading to breast cancer. At molecular level, 87% of STS is expressed and an inhibitor targeting STS could decrease the level of estrogens. In an attempt to identify the chemical structural requirement targeting placental STS inhibition, 26 compounds with pIC50 ranging from 4.61 to 9.46 were subjected to computational studies including Quantitative Structural-Activity Relationship (QSAR), MolecularDocking followed by Density Functional Theory (DFT) studies. A robust and predictable model were developed with good R2 (0.834) and cross-validated correlation coefficient value Q2 LOO (0.786) explaining the relationship quantitatively. The regression graphs suggests that the STS inhibition was greatly dependent on the electro topological state of an atom, sum of the atom type E-state (SdssC), maximum E-states for strong hydrogen bond acceptors (maxHBa) and basic group count descriptor (BCUTp-1h). Furthermore, docking results showed favorable interactions of sulfamate analogs with catalytically important amino acid residues such as LEU74, VAL101, and VAL486. The interactions of the best active compound 3j when compared with standard Irosustat show similar binding energies. DFT studies further confirm the presence of HOMO orbital centered on chromenone ring further highlighting its importance for receptor ligand hydrophobic interaction. The study reveals that substitution of thio in chromenone nucleus and introduction of adamantyl substitution at second position are favorable in inhibiting the enzyme STS.


Asunto(s)
Inhibidores Enzimáticos/química , Antagonistas de Estrógenos/química , Relación Estructura-Actividad Cuantitativa , Esteril-Sulfatasa/ultraestructura , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Inhibidores Enzimáticos/uso terapéutico , Antagonistas de Estrógenos/uso terapéutico , Estrógenos/metabolismo , Femenino , Humanos , Enlace de Hidrógeno/efectos de los fármacos , Simulación del Acoplamiento Molecular , Esteril-Sulfatasa/antagonistas & inhibidores , Esteril-Sulfatasa/química , Esteril-Sulfatasa/genética
8.
J Enzyme Inhib Med Chem ; 35(1): 1163-1184, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32363947

RESUMEN

The purpose of this review article is to provide an overview of recent achievements in the synthesis of novel steroid sulphatase (STS) inhibitors. STS is a crucial enzyme in the biosynthesis of active hormones (including oestrogens and androgens) and, therefore, represents an extremely attractive molecular target for the development of hormone-dependent cancer therapies. The inhibition of STS may effectively reduce the availability of active hormones for cancer cells, causing a positive therapeutic effect. Herein, we report examples of novel STS inhibitors based on steroidal and nonsteroidal cores that contain various functional groups (e.g. sulphamate and phosphorus moieties) and halogen atoms, which may potentially be used in therapies for hormone-dependent cancers. The presented work also includes examples of multitargeting agents with STS inhibitory activities. Furthermore, the fundamental discoveries in the development of the most promising drug candidates exhibiting STS inhibitory activities are highlighted.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Esteril-Sulfatasa/antagonistas & inhibidores , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estructura Molecular , Esteril-Sulfatasa/química , Esteril-Sulfatasa/metabolismo
9.
Bioorg Med Chem ; 28(7): 115368, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32122754

RESUMEN

Steroid sulfatase (STS) is an important enzyme regulating the conversion of sulfated steroids into their active hydroxylated forms. Notably, the inhibition of STS has been shown to decrease the levels of active estrogens and was translated into clinical trials for the treatment of breast cancer. Based on quantitative structure-activity relationship (QSAR) and molecular modeling studies, we herein report the design of fluorescent inhibitors of STS by adding a dansyl group on an estrane scaffold. Synthesis of 17α-dansylaminomethyl-estradiol (7) and its sulfamoylated analog 8 were achieved from estrone in 5 and 6 steps, respectively. Inhibition assays on HEK-293 cells expressing exogenous STS revealed a high level of inhibition for compound 7 (IC50 = 69 nM), a value close to the QSAR model prediction (IC50 = 46 nM). As an irreversible inhibitor, sulfamate 8 led to an even more potent inhibition in the low nanomolar value (IC50 = 2.1 nM). In addition, we show that the potent STS inhibitor 8 can be employed as an optical imaging tool to investigate intracellular enzyme sub-localization as well as inhibitory behavior. As a result, confocal microscopy analysis confirmed good penetration of the STS fluorescent inhibitor 8 in cells and its localization in the endoplasmic reticulum where STS is localized.


Asunto(s)
Estradiol/análogos & derivados , Esteril-Sulfatasa/antagonistas & inhibidores , Estradiol/síntesis química , Estrona/química , Células HEK293 , Humanos , Microscopía Confocal , Modelos Moleculares , Estructura Molecular , Imagen Óptica , Relación Estructura-Actividad Cuantitativa
10.
Bioorg Med Chem ; 28(8): 115406, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32173116

RESUMEN

Steroid sulfatase (STS) has recently emerged as a drug target for management of hormone-dependent malignancies. In the present study, a new series of twenty-one aryl amido-linked sulfamate derivatives 1a-u was designed and synthesized, based upon a cyclohexyl lead compound. All members were evaluated as STS inhibitors in a cell-free assay. Adamantyl derivatives 1h and 1p-r were the most active with more than 90% inhibition at 10 µM concentration and, for those with the greatest inhibitory activity, IC50 values were determined. These compounds exhibited STS inhibition within the range of ca 25-110 nM. Amongst them, compound 1q possessing a o-chlorobenzene sulfamate moiety exhibited the most potent STS inhibitory activity with an IC50 of 26 nM. Furthermore, to assure capability to pass through the cell lipid bilayer, compounds with low IC50 values were tested against STS activity in JEG-3 whole-cell assays. Consequently, 1h and 1q demonstrated IC50 values of ca 14 and 150 nM, respectively. Thus, compound 1h is 31 times more potent than the corresponding cyclohexyl lead (IC50 value = 421 nM in a JEG-3 whole-cell assay). Furthermore, the most potent STS inhibitors (1h and 1p-r) were evaluated for their antiproliferative activity against the estrogen-dependent breast cancer cell line T-47D. They showed promising activity with single digit micromolar IC50 values (ca 1-6 µM) and their potency against T-47D cells was comparable to that against STS enzyme. In conclusion, this new class of adamantyl-containing aryl sulfamate inhibitor has potential for further development against hormone-dependent tumours.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Ácidos Sulfónicos/química , Antineoplásicos/química , Neoplasias de la Mama , Sistema Libre de Células , Femenino , Humanos , Concentración 50 Inhibidora , Estructura Molecular , Esteril-Sulfatasa/antagonistas & inhibidores , Relación Estructura-Actividad
11.
Bioorg Chem ; 96: 103618, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32059152

RESUMEN

Steroid sulfatase (STS) is a sulfatase enzyme that catalyzes the conversion of sulfated steroid precursors to free steroid. The inhibition of STS could abate estrogenic steroids that stimulate the proliferation and development of breast cancer, and therefore STS is a potential target for adjuvant endocrine therapy. In this study, a series of 3-benzylaminocoumarin-7-O-sulfamate derivatives targeting STS were designed and synthesized. Structure-relationship activities (SAR) analysis revealed that attachment of a benzylamino group at the 3-position of coumarin improved inhibitory activity. Compound 3j was found to have the highest inhibition activity against human placenta isolated STS (IC50  0.13 µM) and MCF-7 cell lines (IC50 1.35 µM). Kinetic studies found compound 3j to be an irreversible inhibitor of STS, with KI and kinact value of 86.9 nM and 158.7 min-1, respectively.


Asunto(s)
Cumarinas/química , Cumarinas/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Esteril-Sulfatasa/antagonistas & inhibidores , Aminación , Compuestos de Bencilo/síntesis química , Compuestos de Bencilo/química , Compuestos de Bencilo/farmacología , Cumarinas/síntesis química , Inhibidores Enzimáticos/síntesis química , Femenino , Humanos , Células MCF-7 , Placenta/enzimología , Embarazo , Esteril-Sulfatasa/metabolismo , Relación Estructura-Actividad , Ácidos Sulfónicos/síntesis química , Ácidos Sulfónicos/química , Ácidos Sulfónicos/farmacología
12.
Biol Futur ; 71(3): 249-264, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34554507

RESUMEN

The potential inhibitory effect of diverse triazolyl-ferrocene steroids on key enzymes of the estrogen biosynthesis was investigated. Test compounds were synthesized via copper-catalyzed cycloaddition of steroidal azides and ferrocenyl-alkynes using our efficient methodology published previously. Inhibition of human aromatase, steroid sulfatase (STS) and 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD1) activities was investigated with in vitro radiosubstrate incubations. Some of the test compounds were found to be potent inhibitors of the STS. A compound bearing ferrocenyl side chain on the C-2 displayed a reversible inhibition, whereas C-16 and C-17 derivatives displayed competitive irreversible binding mechanism toward the enzyme. 17α-Triazolyl-ferrocene derivatives of 17ß-estradiol exerted outstanding inhibitory effect and experiments demonstrated a key role of the ferrocenyl moiety in the enhanced binding affinity. Submicromolar IC50 and Ki parameters enroll these compounds to the group of the most effective STS inhibitors published so far. STS inhibitory potential of the steroidal ferrocenes may lead to the development of novel compounds able to suppress in situ biosynthesis of 17ß-estradiol in target tissues.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , Inhibidores de la Aromatasa/síntesis química , Compuestos Ferrosos/química , Metalocenos/química , Esteril-Sulfatasa/antagonistas & inhibidores , Triazoles/química , Estrógenos/biosíntesis
13.
Bioorg Chem ; 95: 103495, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31855822

RESUMEN

Steroid sulfatase (STS) transforms hormone precursors into active steroids. Thus, it represents a target of intense research regarding hormone-dependent cancers. In this study, three ligand-based pharmacophore models were developed to identify STS inhibitors from natural sources. In a pharmacophore-based virtual screening of a curated molecular TCM database, lanostane-type triterpenes (LTTs) were predicted as STS ligands. Three traditionally used polypores rich in LTTs, i.e., Ganoderma lucidum Karst., Gloeophyllum odoratum Imazeki, and Fomitopsis pinicola Karst., were selected as starting materials. Based on eighteen thereof isolated LTTs a structure activity relationship for this compound class was established with piptolinic acid D (1), pinicolic acid B (2), and ganoderol A (3) being the most pronounced and first natural product STS inhibitors with IC50 values between 10 and 16 µM. Molecular docking studies proposed crucial ligand target interactions and a prediction tool for these natural compounds correlating with experimental findings.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Lanosterol/farmacología , Esteril-Sulfatasa/antagonistas & inhibidores , Triterpenos/farmacología , Basidiomycota/química , Coriolaceae/química , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/aislamiento & purificación , Humanos , Lanosterol/análogos & derivados , Lanosterol/química , Ligandos , Modelos Moleculares , Estructura Molecular , Reishi/química , Esteril-Sulfatasa/metabolismo , Relación Estructura-Actividad , Triterpenos/química , Triterpenos/aislamiento & purificación
14.
Eur J Med Chem ; 182: 111614, 2019 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-31422224

RESUMEN

Two new piperazinyl-ureido single ring aryl sulfamate-based inhibitor series were designed against the emerging oncology drug target steroid sulfatase (STS), for which there are existing potent steroidal and non-steroidal agents in clinical trials. 4-(Piperazinocarbonyl)aminosulfamates (5-31) were obtained by reacting 4-hydroxyarylamines with phenylchloroformate, subsequent sulfamoylation of the resulting hydroxyarylcarbamates and coupling of the product with 1-substituted piperazines. Pyrimidinyl-piperazinourea sulfamates (35-42) were synthesized by pyrimidine ring closure of 4-Boc-piperazine-1-carboxamidine with 3-(dimethylamino)propenones, deprotection and coupling with the sulfamoylated building block. Target ureidosulfamates 5-31 and 35-42 were evaluated both as STS inhibitors in vitro using a lysate of JEG-3 human placenta choriocarcinoma cell line and in a whole cell assay. SAR conclusions were drawn from both series. In series 35-42 the best inhibitory activity is related to the presence of a benzofuryl on the pyrimidine ring. In series 5-31 the best inhibitory activity was shown by the ureas bearing 4-chlorophenyl, 3,4-dichlorophenyl groups or aliphatic chains at the piperazino 4-nitrogen displaying IC50 in the 33-94 nM concentration range. Final optimization to the low nanomolar level was achieved through substitution of the arylsulfamate ring with halogens. Four halogenated arylsulfamates of high potency were achieved and two of these 19 and 20 had IC50 values of 5.1 and 8.8 nM respectively and are attractive for potential in vivo evaluation and further development. We demonstrate the optimization of this new series to low nanomolar potency, employing fluorine substitution, providing potent membrane permeant inhibitors with further development potential indicating piperazinyl-ureido aryl sulfamate derivatives as an attractive new class of STS inhibitors.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Piperazinas/farmacología , Esteril-Sulfatasa/antagonistas & inhibidores , Ácidos Sulfónicos/farmacología , Urea/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Estructura Molecular , Piperazinas/síntesis química , Piperazinas/química , Esteril-Sulfatasa/metabolismo , Relación Estructura-Actividad , Ácidos Sulfónicos/síntesis química , Ácidos Sulfónicos/química , Urea/análogos & derivados , Urea/química
15.
Drug Dev Res ; 80(6): 857-866, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31301181

RESUMEN

In the present work, we described convenient methods for the synthesis of N-thiophosphorylated 3-(4-aminophenyl)-coumarin-7-O-sulfamates as steroid sulfatase (STS) inhibitors. To design the structures of the potential STS inhibitors, molecular modeling techniques were used. A computational docking method was used to determine the binding modes of the synthesized inhibitors as well as to identify potential interactions between specified functional groups on the inhibitors and the amino acid residues present in the active site of the enzyme. The inhibitory activities of the synthesized compounds were tested in an enzymatic assay with STS isolated from a human placenta. Within the set of newly synthesized compounds, 9e demonstrated the highest inhibitory activity in the enzymatic assay with an IC50 value of 0.201 µM (the IC50 value of 667-COUMATE in the same test was 0.062 µM). Furthermore, we tried to verify if the obtained STS inhibitors are able to pass through the cellular membrane effectively in cell line experiments. In the course of our study, we determined the STS activity in the MCF-7 cell line after incubation in the presence of the inhibitors (at 100 nM concentration). For this evaluation, we included newly synthesized compounds 9a-g and their N-phosphorylated analogs 6a-h, whose synthesis has been previously described. We found that the lowest STS activities were measured in the presence of N-phosphorylated derivatives 6e (0.1% of STS activity) and 6f (0.2% of STS activity). The measured STS activity in the presence of 667-COUMATE (used as a reference) was 0.1%. Moreover, at concentrations up to 1 µM, the most active compounds (6e, 6f, 9b, and 9e) did not exert any toxic effects on zebrafish embryos.


Asunto(s)
Cumarinas/farmacología , Esteril-Sulfatasa/antagonistas & inhibidores , Sulfonamidas/farmacología , Animales , Cumarinas/química , Embrión no Mamífero , Humanos , Células MCF-7 , Simulación del Acoplamiento Molecular , Esteril-Sulfatasa/química , Esteril-Sulfatasa/metabolismo , Sulfonamidas/química , Pez Cebra
16.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2464-2474, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31195119

RESUMEN

Human steroid sulfatase (STS) has been linked with poor prognosis in steroid-associated tumors and represents an important clinical target in cancers, yet the mechanism of STS-induced carcinogenesis remains unclear. To correlate STS with cancer metabolism, we determined the effects of STS on aerobic glycolysis. STS overexpression increased cellular levels of lactic acid, the final product of aerobic glycolysis. Moreover, STS suppressed the oxygen consumption rate (OCR), which represents mitochondrial respiration. Inhibition of STS by the specific inhibitor STX064 recovered STS-induced OCR repression and lactic acid over-production. DHEA, but not DHEA-S, suppressed the OCR level and enhanced lactic acid production. To understand the molecular mechanism of STS-induced cancer metabolism, we measured the expression of glycolytic enzymes hexokinase 2 (HK2) and pyruvate kinase M2 (PKM2), which was highly upregulated by STS and DHEA at both protein and mRNA levels. HIF1α is a key mediator of aerobic glycolysis, and STS enhanced HIF1α promoter activity, mRNA expression, and protein expression. Down-regulation of HIF1α by siRNA suppressed the HK2 and PKM2 expression induced by both STS and DHEA. HIF1α siRNA also recovered the OCR repression and lactic acid over-production induced by both STS and DHEA. To explore the mechanism in vivo, we produced transgenic mice overexpressing STS and found that STS expression was particularly enhanced in the lung. Consistent with our in vitro results, the expression of HIF1α, HK2, and PKM2 was also increased in mouse lung tissues. In conclusion, we suggest that STS may induce aerobic glycolysis through enhancing HIF1α expression.


Asunto(s)
Proteínas Portadoras/metabolismo , Glucólisis , Hexoquinasa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas de la Membrana/metabolismo , Esteril-Sulfatasa/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Proteínas Portadoras/genética , Deshidroepiandrosterona/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Glucólisis/efectos de los fármacos , Células HeLa , Hexoquinasa/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Pulmón/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Consumo de Oxígeno/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Esteril-Sulfatasa/antagonistas & inhibidores , Hormonas Tiroideas/genética , Proteínas de Unión a Hormona Tiroide
17.
J Steroid Biochem Mol Biol ; 193: 105411, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31207361

RESUMEN

Reductive 17ß-hydroxysteroid dehydrogenases (17ß-HSDs) and 11ß-hydroxysteroid dehydrogenase 2 (11ß-HSD2) play crucial roles in respectively regulating steroids and glucocorticoids for the progression of hormone-dependent breast cancer. Most studies focused on the function and individual regulation of these enzymes. However, mutual regulation of these enzymes and the induced modulation on the estrogen and androgen receptors for breast cancer promotion are not yet clear. In this study, MCF-7 and T47D cells were treated with inhibitors of 17ß-HSD1, 17ß-HSD7, aromatase or steroid sulfatase (STS), then mRNA levels of 17ß-HSD7, STS, 11ß-HSD 2, estrogen receptors α (ERα) and androgen receptor (AR) were determined by Q-PCR. ER negative cell line MDA-MB-231 was used as a negative control. Our results demonstrate that 17ß-HSD7, STS and 11ß-HSD2 are all regulated by the same estrogen estradiol via ERα. When the gene of ERα (ESR1) was knocked down, there was no longer significant mutual regulation of these enzymes. Our results demonstrate that important steroidogenic enzymes transcriptionally regulated by ERα, can be mutually closely correlated. Inhibition of one of them can reduce the expression of another, thereby amplifying the role of the inhibition. Furthermore, inhibition of 17ß-HSD7 increases the expression of AR gene which is considered as a marker for better prognosis in ER + breast cancer, while maintaining ERα level. Thus, our mechanistic finding provides a base for further improving the endocrine therapy of ER + breast cancer, e.g., for selecting the target steroid enzymes, and for the combined targeting of human 17ß-HSD7 and ERα.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/genética , Receptores Androgénicos/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/antagonistas & inhibidores , 17-Hidroxiesteroide Deshidrogenasas/genética , Inhibidores de la Aromatasa/farmacología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Dihidrotestosterona/metabolismo , Estradiol/metabolismo , Femenino , Humanos , Esteril-Sulfatasa/antagonistas & inhibidores , Esteril-Sulfatasa/genética , Esteril-Sulfatasa/metabolismo
18.
J Biol Chem ; 294(32): 12112-12121, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31217279

RESUMEN

Conjugated equine estrogens (CEEs), whose brand name is Premarin, are widely used as a hormone-replacement therapy (HRT) drug to manage postmenopausal symptoms in women. Extracted from pregnant mare urine, CEEs are composed of nearly a dozen estrogens existing in an inactive sulfated form. To determine whether the hepatic steroid sulfatase (STS) is a key contributor to the efficacy of CEEs in HRT, we performed estrogen-responsive element (ERE) reporter gene assay, real-time PCR, and UPLC-MS/MS to assess the STS-dependent and inflammation-responsive estrogenic activity of CEEs in HepG2 cells and human primary hepatocytes. Using liver-specific STS-expressing transgenic mice, we also evaluated the effect of STS on the estrogenic activity of CEEs in vivo We observed that CEEs induce activity of the ERE reporter gene in an STS-dependent manner and that genetic or pharmacological inhibition of STS attenuates CEE estrogenic activity. In hepatocytes, inflammation enhanced CEE estrogenic activity by inducing STS gene expression. The inflammation-responsive estrogenic activity of CEEs, in turn, attenuated inflammation through the anti-inflammatory activity of the active estrogens. In vivo, transgenic mice with liver-specific STS expression exhibited markedly increased sensitivity to CEE-induced estrogenic activity in the uterus resulting from increased levels of liver-derived and circulating estrogens. Our results reveal a critical role of hepatic STS in mediating the hormone-replacing activity of CEEs. We propose that caution needs to be applied when Premarin is used in patients with chronic inflammatory liver diseases because such patients may have heightened sensitivity to CEEs due to the inflammatory induction of STS activity.


Asunto(s)
Estrógenos Conjugados (USP)/metabolismo , Esteril-Sulfatasa/metabolismo , Animales , Células Cultivadas , Cromatografía Líquida de Alta Presión , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Estrógenos Conjugados (USP)/análisis , Estrógenos Conjugados (USP)/farmacología , Femenino , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Caballos , Humanos , Inflamación/metabolismo , Inflamación/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Esteril-Sulfatasa/antagonistas & inhibidores , Esteril-Sulfatasa/genética , Espectrometría de Masas en Tándem , Útero/efectos de los fármacos , Útero/metabolismo , Útero/patología
19.
J Med Chem ; 62(11): 5512-5521, 2019 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-31062594

RESUMEN

Steroid sulfatase (STS) is a key enzyme involved in the biosynthesis of estrogens from inactive sulfated steroids. After we reported EO-33 as a potent in vitro STS inhibitor without undesirable estrogenic activity and with osteogenic properties, we are now interested in validating EO-33's in vivo potential to inhibit STS, to prevent bone deterioration, and to reduce estrogen-dependent tumor growth. A scale-up synthesis was first elaborated to prepare the multigram quantity of EO-33 needed to perform in vivo studies. EO-33 blocked the uterine weight stimulated by estrone sulfate in ovariectomized mice by 69% and the STS activity in the liver by 81%. It also produced a selective estrogen receptor modulator effect as assessed by measuring the tibia weight and calcium content. Using a human breast cancer (MCF-7 xenograft) model in nude mice, EO-33 blocked 90% of tumor growth induced by estradiol sulfate, and no toxic effect was observed by assessing the body and liver weights.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Osteogénesis/efectos de los fármacos , Esteril-Sulfatasa/antagonistas & inhibidores , Ácidos Sulfónicos/farmacología , Animales , Antineoplásicos/química , Inhibidores Enzimáticos/química , Estrógenos/biosíntesis , Humanos , Células MCF-7 , Ratones , Ácidos Sulfónicos/química , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Steroid Biochem Mol Biol ; 185: 110-117, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30118815

RESUMEN

Steroid sulfatase (STS) plays an important role in the regulation of steroid hormones. Metabolism of steroid hormones in zebrafish has been investigated, but the action of steroid sulfatase remains unknown. In this study, a zebrafish sts was cloned, expressed, purified, and characterized in comparison with the orthologous human enzyme. Enzymatic assays demonstrated that similar to human STS, zebrafish Sts was most active in catalyzing the hydrolysis of estrone-sulfate and estradiol-sulfate, among five steroid sulfates tested as substrates. Kinetic analyses revealed that the Km values of zebrafish Sts and human STS differed with respective substrates, but the catalytic efficiency as reflected by the Vmax/Km appeared comparable, except for DHEA-sulfate with which zebrafish Sts appeared less efficient. While zebrafish Sts was catalytically active at 28 °C, the enzyme appeared more active at 37 °C and with similar Km values to those determined at 28 °C. Assays performed in the presence of different divalent cations showed that the activities of both zebrafish and human STSs were stimulated by Ca2+, Mg2+, and Mn2+, and inhibited by Zn+2 and Fe2+. EMATE and STX64, two known mammalian steroid sulafatase inhibitors, were shown to be capable of inhibiting the activity of zebrafish Sts. Collectively, the results obtained indicated that zebrafish Sts exhibited enzymatic characteristics comparable to the human STS, suggesting that the physiological function of STS may be conserved between zebrafish and humans.


Asunto(s)
Sulfato de Deshidroepiandrosterona/metabolismo , Estradiol/análogos & derivados , Estrona/análogos & derivados , Esteril-Sulfatasa/genética , Esteril-Sulfatasa/metabolismo , Secuencia de Aminoácidos , Animales , Catálisis , Cationes/metabolismo , Clonación Molecular/métodos , Inhibidores Enzimáticos/farmacología , Estradiol/metabolismo , Estrona/metabolismo , Humanos , Esteril-Sulfatasa/antagonistas & inhibidores , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...