Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
ACS Omega ; 3(9): 10748-10772, 2018 Sep 30.
Article in English | MEDLINE | ID: mdl-30320251

ABSTRACT

Synthetic routes to potent bicyclic nonsteroidal sulfamate-based active-site-directed inhibitors of the enzyme steroid sulfatase (STS), an emerging target in the treatment of postmenopausal hormone-dependent diseases, including breast cancer, are described. Sulfamate analogs 9-27 and 28-46 of the core in vivo active two-ring coumarin template, modified at the 4- and 3-positions, respectively, were synthesized to expand structure-activity relationships. α-Alkylacetoacetates were used to synthesize coumarin sulfamate derivatives with 3-position modifications, and the bicyclic ring of other parent coumarins was primarily constructed via the Pechmann synthesis of hydroxyl coumarins. Compounds were examined for STS inhibition in intact MCF-7 breast cancer cells and in placental microsomes. Low nanomolar potency STS inhibitors were achieved, and some were found to inhibit the enzyme in MCF-7 cells ca. 100-500 more potently than the parent 4-methylcoumarin-7-O-sulfamate 3, with the best compounds close in potency to the tricyclic clinical drug Irosustat. 3-Hexyl-4-methylcoumarin-7-O-sulfamate 29 and 3-benzyl-4-methylcoumarin-7-O-sulfamate 41 were particularly effective inhibitors with IC50 values of 0.68 and 1 nM in intact MCF-7 cells and 8 and 32 nM for placental microsomal STS, respectively. They were docked into the STS active site for comparison with estrone 3-O-sulfamate and Irosustat, showing their sulfamate group close to the catalytic hydrated formylglycine residue and their pendant group lying between the hydrophobic sidechains of L103, F178, and F488. Such highly potent STS inhibitors expand the structure-activity relationship for these coumarin sulfamate-based agents that possess therapeutic potential and may be worthy of further development.

2.
ChemMedChem ; 8(5): 779-99, 2013 May.
Article in English | MEDLINE | ID: mdl-23495205

ABSTRACT

4-{[(4-Cyanophenyl)(4H-1,2,4-triazol-4-yl)amino]methyl}phenyl sulfamate and its ortho-halogenated (F, Cl, Br) derivatives are first-generation dual aromatase and sulfatase inhibitors (DASIs). Structure-activity relationship studies were performed on these compounds, and various modifications were made to their structures involving relocation of the halogen atom, introduction of more halogen atoms, replacement of the halogen with another group, replacement of the methylene linker with a difluoromethylene linker, replacement of the para-cyanophenyl ring with other ring structures, and replacement of the triazolyl group with an imidazolyl group. The most potent in vitro DASI discovered is an imidazole derivative with IC50 values against aromatase and steroid sulfatase in a JEG-3 cell preparation of 0.2 and 2.5 nM, respectively. The parent phenol of this compound inhibits aromatase with an IC50 value of 0.028 nM in the same assay.


Subject(s)
Aromatase/metabolism , Enzyme Inhibitors/pharmacology , Steryl-Sulfatase/antagonists & inhibitors , Sulfonamides/pharmacology , Triazoles/pharmacology , Cell Line , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Steryl-Sulfatase/metabolism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Triazoles/chemical synthesis , Triazoles/chemistry
3.
ChemMedChem ; 6(11): 2019-34, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-21990014

ABSTRACT

Structure-activity relationship studies were conducted on Irosustat (STX64, BN83495), the first steroid sulfatase (STS) inhibitor to enter diverse clinical trials for patients with advanced hormone-dependent cancer. The size of its aliphatic ring was expanded; its sulfamate group was N,N-dimethylated, relocated to another position and flanked by an adjacent methoxy group; and series of quinolin-2(1H)-one and quinoline derivatives of Irosustat were explored. The STS inhibitory activities of the synthesised compounds were assessed in a preparation of JEG-3 cells. Stepwise enlargement of the aliphatic ring from 7 to 11 members increases potency, although a further increase in ring size is detrimental. The best STS inhibitors in vitro had IC50 values between 0.015 and 0.025 nM. Other modifications made to Irosustat were found to either abolish or significantly weaken its activity. An azomethine adduct of Irosustat with N,N-dimethylformamide (DMF) was isolated, and crystal structures of Irosustat and this adduct were determined. Docking studies were conducted to explore the potential interactions between compounds and the active site of STS, and suggest a sulfamoyl group transfer to formylglycine 75 during the inactivation mechanism.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Steryl-Sulfatase/antagonists & inhibitors , Sulfonic Acids/chemistry , Sulfonic Acids/pharmacology , Azo Compounds/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Dimethylformamide , Drug Evaluation, Preclinical/methods , Formamides/chemistry , Humans , Microsomes/drug effects , Microsomes/enzymology , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Thiosemicarbazones/chemistry
4.
Mol Cell Endocrinol ; 340(2): 154-60, 2011 Jul 04.
Article in English | MEDLINE | ID: mdl-21693170

ABSTRACT

Steroid sulfatase plays a pivotal role in regulating the formation of biologically active steroids from inactive steroid sulfates. It is responsible for the hydrolysis of estrone sulfate and dehydroepiandrosterone sulfate to estrone and dehydroepiandrosterone, respectively, both of which can be subsequently reduced to steroids with estrogenic properties (i.e. estradiol and androstenediol) that can stimulate the growth of tumors in hormone-responsive tissues of the breast, endometrium and prostate. Hence, the action of steroid sulfatase is implicated in physiological processes and pathological conditions. It has been five years since our group last reviewed the important role of this enzyme in steroid synthesis and the progress made in the development of potent inhibitors of this important enzyme target. This timely review therefore concentrates on recent advances in steroid sulfatase research, and summarises the findings of clinical trials with Irosustat (BN83495), the only steroid sulfatase inhibitor that is being trialed in postmenopausal women with breast or endometrial cancer.


Subject(s)
Estrogens/biosynthesis , Estrogens/metabolism , Steryl-Sulfatase/metabolism , Clinical Trials as Topic , Enzyme Inhibitors/pharmacology , Humans , Steryl-Sulfatase/antagonists & inhibitors
5.
ChemMedChem ; 6(8): 1423-38, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21608133

ABSTRACT

Concurrent inhibition of aromatase and steroid sulfatase (STS) may provide a more effective treatment for hormone-dependent breast cancer than monotherapy against individual enzymes, and several dual aromatase-sulfatase inhibitors (DASIs) have been reported. Three aromatase inhibitors with sub-nanomolar potency, better than the benchmark agent letrozole, were designed. To further explore the DASI concept, a new series of letrozole-derived sulfamates and a vorozole-based sulfamate were designed and biologically evaluated in JEG-3 cells to reveal structure-activity relationships. Amongst achiral and racemic compounds, 2-bromo-4-(2-(4-cyanophenyl)-2-(1H-1,2,4-triazol-1-yl)ethyl)phenyl sulfamate is the most potent DASI (aromatase: IC50 =0.87 nM; STS: IC50 =593 nM). The enantiomers of the phenolic precursor to this compound were separated by chiral HPLC and their absolute configuration determined by X-ray crystallography. Following conversion to their corresponding sulfamates, the S-(+)-enantiomer was found to inhibit aromatase and sulfatase most potently (aromatase: IC50 =0.52 nM; STS: IC50 =280 nM). The docking of each enantiomer and other ligands into the aromatase and sulfatase active sites was also investigated.


Subject(s)
Aromatase Inhibitors/chemistry , Aromatase/chemistry , Nitriles/chemistry , Steryl-Sulfatase/antagonists & inhibitors , Triazoles/chemistry , Aromatase/metabolism , Aromatase Inhibitors/chemical synthesis , Aromatase Inhibitors/pharmacology , Binding Sites , Catalytic Domain , Cell Line, Tumor , Computer Simulation , Crystallography, X-Ray , Enzyme Activation/drug effects , Humans , Letrozole , Nitriles/pharmacology , Stereoisomerism , Steryl-Sulfatase/metabolism , Structure-Activity Relationship , Sulfonic Acids/chemistry , Sulfonic Acids/pharmacology , Triazoles/pharmacology
6.
Mol Cell Endocrinol ; 340(2): 175-85, 2011 Jul 04.
Article in English | MEDLINE | ID: mdl-21238537

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.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Steryl-Sulfatase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Estrone/analogs & derivatives , Estrone/pharmacology , Humans , Steryl-Sulfatase/metabolism , Sulfonic Acids/chemistry , Sulfonic Acids/pharmacology
7.
ACS Med Chem Lett ; 2(3): 243-7, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-24900302

ABSTRACT

Single agents against multiple drug targets are highly topical. Hormone-dependent breast cancer (HDBC) may be more effectively treated by dual inhibition of aromatase and steroid sulfatase (STS), and several dual aromatase-sulfatase inhibitors (DASIs) have been recently reported. The best compounds from two leading classes of DASI, 3 and 9, are low nanomolar inhibitors. In search of a novel class of DASI, core motifs of two leading classes were combined to give a series of hybrid structures, with several compounds showing markedly improved dual inhibitory activities in the picomolar range in JEG-3 cells. Thus, DASIs 14 (IC50: aromatase, 15 pM; STS, 830 pM) and 15 (IC50: aromatase, 18 pM; STS, 130 pM) are the first examples of an exceptional new class of highly potent dual inhibitor that should encourage further development toward multitargeted therapeutic intervention in HDBC.

8.
ChemMedChem ; 5(9): 1577-93, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20632362

ABSTRACT

The design and synthesis of a series of bicyclic ring containing dual aromatase-sulfatase inhibitors (DASIs) based on the aromatase inhibitor (AI) 4-[(4-bromobenzyl)(4H-1,2,4-triazol-4-yl)amino]benzonitrile are reported. Biological evaluation with JEG-3 cells revealed structure-activity relationships. The X-ray crystal structure of sulfamate 23 was determined, and selected compounds were docked into the aromatase and steroid sulfatase (STS) crystal structures. In the sulfamate-containing series, compounds containing a naphthalene ring are both the most potent AI (39, IC(50 AROM)=0.25 nM) and the best STS inhibitor (31, IC(50 STS)=26 nM). The most promising DASI is 39 (IC(50 AROM)=0.25 nM, IC(50 STS)=205 nM), and this was evaluated orally in vivo at 10 mg kg(-1), showing potent inhibition of aromatase (93 %) and STS (93 %) after 3 h. Potent aromatase and STS inhibition can thus be achieved with a DASI containing a bicyclic ring system; development of such a DASI could provide an attractive new option for the treatment of hormone-dependent breast cancer.


Subject(s)
Antineoplastic Agents/chemical synthesis , Aromatase Inhibitors/chemical synthesis , Aromatase/chemistry , Steryl-Sulfatase/antagonists & inhibitors , Sulfonic Acids/chemistry , Triazoles/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Aromatase/metabolism , Aromatase Inhibitors/chemistry , Aromatase Inhibitors/pharmacology , Binding Sites , Cell Line, Tumor , Computer Simulation , Crystallography, X-Ray , Humans , Molecular Conformation , Steryl-Sulfatase/metabolism , Structure-Activity Relationship , Sulfonic Acids/chemical synthesis , Sulfonic Acids/pharmacology , Triazoles/chemistry , Triazoles/pharmacology
9.
J Med Chem ; 53(5): 2155-70, 2010 Mar 11.
Article in English | MEDLINE | ID: mdl-20148564

ABSTRACT

Single agents against multiple drug targets are of increasing interest. Hormone-dependent breast cancer (HDBC) may be more effectively treated by dual inhibition of aromatase and steroid sulfatase (STS). The aromatase inhibitory pharmacophore was thus introduced into a known biphenyl STS inhibitor to give a series of novel dual aromatase-sulfatase inhibitors (DASIs). Several compounds are good aromatase or STS inhibitors and DASI 20 (IC(50): aromatase, 2.0 nM; STS, 35 nM) and its chlorinated congener 23 (IC(50): aromatase, 0.5 nM; STS, 5.5 nM) are examples that show exceptional dual potency in JEG-3 cells. Both biphenyls share a para-sulfamate-containing ring B and a ring A, which contains a triazol-1-ylmethyl meta to the biphenyl bridge and para to a nitrile. At 1 mg/kg po, 20 and 23 reduced plasma estradiol levels strongly and inhibited liver STS activity potently in vivo. 23 is nonestrogenic and potently inhibits carbonic anhydrase II (IC(50) 86 nM). A complex was crystallized and its structure was solved by X-ray crystallography. This class of DASI should encourage further development toward multitargeted therapeutic intervention in HDBC.


Subject(s)
Aromatase Inhibitors/chemistry , Aromatase Inhibitors/pharmacology , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Steryl-Sulfatase/antagonists & inhibitors , Triazoles/chemistry , Triazoles/pharmacology , Animals , Aromatase Inhibitors/chemical synthesis , Biphenyl Compounds/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Female , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Rats , Rats, Wistar , Spectrometry, Mass, Electrospray Ionization , Steryl-Sulfatase/metabolism , Structure-Activity Relationship , Triazoles/chemical synthesis
10.
Ann N Y Acad Sci ; 1155: 80-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19250195

ABSTRACT

Steroid sulfatase (STS) regulates the hydrolysis of steroid sulfates to their unconjugated forms. Estrone sulfate and dehydroepiandrosterone sulfate can be hydrolyzed by STS to estrone and dehydroepiandrosterone, respectively, with these steroids being the precursors for the synthesis of more biologically active estrogens or androgens. A number of potent STS inhibitors have now been developed including STX64, which entered a phase I trial for the treatment of postmenopausal women with advanced metastatic hormone-dependent breast cancer. The results from this phase I trial were encouraging, suggesting that STS inhibitors may also have a role in the treatment of other hormone-dependent cancers including those of the endometrium, ovary, and prostate. In this paper the potential use of STS inhibitors to treat these hormone-dependent cancers is reviewed. In addition, results from in vitro studies show that Ishikawa endometrial cancer cells, OVCAR-3 ovarian cancer cells, and LNCaP prostate cancer cells all possess significant STS activity. Furthermore, STS activity in these cells can be almost completely inhibited by STX64 or the second-generation STS inhibitor, STX213. Results from these investigations therefore suggest that STS inhibitors could have therapeutic potential for the treatment of a range of hormone-dependent cancers.


Subject(s)
Enzyme Inhibitors/chemistry , Neoplasms, Hormone-Dependent/drug therapy , Steryl-Sulfatase/antagonists & inhibitors , Breast Neoplasms/drug therapy , Cell Line, Tumor , Endometrial Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Prostatic Neoplasms/drug therapy , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Steryl-Sulfatase/genetics
11.
Clin Cancer Res ; 14(20): 6469-77, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18927286

ABSTRACT

PURPOSE: The production of E2 is paramount for the growth of estrogen receptor-positive breast cancer. Various strategies have been used, including the use of enzyme inhibitors against either aromatase (AROM) or steroid sulfatase (STS), in an attempt to ablate E2 levels. Both these enzymes play a critical role in the formation of estrogenic steroids and their inhibitors are now showing success in the clinic. EXPERIMENTAL DESIGN: We show here, in a xenograft nude mouse model, that the inhibition of both enzymes using STX681, a dual AROM and STS inhibitor (DASI), is a potential new therapeutic strategy against HDBC. MCF-7 cells stably expressing either AROM cDNA (MCF-7(AROM)) or STS cDNA (MCF-7(STS)) were generated. Ovariectomized MF-1 female nude mice receiving s.c. injections of either androstenedione (A(4)) or E2 sulfate and bearing either MCF-7(AROM) or MCF-7(STS) tumors were orally treated with STX64, letrozole, or STX681. Treatment was administered for 28 days. Mice were weighed and tumor measurements were taken weekly. RESULTS: STX64, a potent STS inhibitor, completely blocked MCF-7(STS) tumor growth but failed to attenuate MCF-7(AROM) tumor growth. In contrast, letrozole inhibited MCF-7(AROM) tumors but had no effect on MCF-7(STS) tumors. STX681 completely inhibited the growth of both tumors. AROM and STS activity was also completely inhibited by STX681, which was accompanied by a significant reduction in plasma E2 levels. CONCLUSIONS: This study indicates that targeting both the AROM and the STS enzyme with a DASI inhibits HDBC growth and is therefore a potentially novel treatment for this malignancy.


Subject(s)
Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Steryl-Sulfatase/antagonists & inhibitors , Administration, Oral , Animals , Azasteroids/therapeutic use , Breast Neoplasms/enzymology , Breast Neoplasms/surgery , Cell Proliferation/drug effects , Estrogens/blood , Female , Humans , Letrozole , Mice , Mice, Nude , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/surgery , Nitriles/therapeutic use , Ovariectomy , Rats , Rats, Wistar , Steryl-Sulfatase/metabolism , Treatment Outcome , Triazoles/therapeutic use , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
ChemMedChem ; 3(11): 1708-30, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18816537

ABSTRACT

4-(((4-Cyanophenyl)(4H-1,2,4-triazol-4-yl)amino)methyl)phenyl sulfamate (6 a) was the first dual aromatase-sulfatase inhibitor (DASI) reported. Several series of its derivatives with various linker systems between the steroid sulfatase (STS) and the aromatase inhibitory pharmacophores were synthesised and evaluated in JEG-3 cells. The X-ray crystal structures of the aromatase inhibitors, DASI precursors 42 d and 60, and DASI 43 h were determined. Nearly all derivatives show improved in vitro aromatase inhibition over 6 a but decreased STS inhibition. The best aromatase inhibitor is 42 e (IC(50)=0.26 nM) and the best DASI is 43 e (IC(50 aromatase)=0.45 nM, IC(50 STS)=1200 nM). SAR for aromatase inhibition shows that compounds containing an alkylene- and thioether-based linker system are more potent than those that are ether-, sulfone-, or sulfonamide-based, and that the length of the linker has a limited effect on aromatase inhibition beyond two methylene units. Compounds 43 d-f were studied in vivo (10 mg kg(-1), single, p.o.). The most potent DASI is 43 e, which inhibited PMSG-induced plasma estradiol levels by 92 % and liver STS activity by 98 % 3 h after dosing. These results further strengthen the concept of designing and developing DASIs for potential treatment of hormone-related cancers.


Subject(s)
Aromatase Inhibitors/chemical synthesis , Aromatase Inhibitors/pharmacology , Chemistry, Pharmaceutical/methods , Nitriles/chemistry , Steryl-Sulfatase/antagonists & inhibitors , Triazoles/chemistry , Amino Acid Motifs , Aromatase/metabolism , Aromatase Inhibitors/chemistry , Cell Line, Tumor , Crystallography, X-Ray/methods , Drug Design , Estradiol/chemistry , Humans , In Vitro Techniques , Inhibitory Concentration 50 , Molecular Conformation , Structure-Activity Relationship , Sulfonamides/chemistry
13.
Mol Cancer Ther ; 7(8): 2435-44, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18723489

ABSTRACT

An improved steroid sulfatase inhibitor was prepared by replacing the N-propyl group of the second-generation steroid-like inhibitor (2) with a N-3,3,3-trifluoropropyl group to give (10). This compound is 5-fold more potent in vitro, completely inhibits rat liver steroid sulfatase activity after a single oral dose of 0.5 mg/kg, and exhibits a significantly longer duration of inhibition over (2). These biological properties are attributed to the increased lipophilicity and metabolic stability of (10) rendered by its trifluoropropyl group and also the potential H-bonding between its fluorine atom(s) and Arg(98) in the active site of human steroid sulfatase. Like other sulfamates, (10) is expected to be sequestered, and transported by, erythrocytes in vivo because it inhibits human carbonic anhydrase II (hCAII) potently (IC(50), 3 nmol/L). A congener (4), which possesses a N-(pyridin-3-ylmethyl) substituent, is even more active (IC(50), 0.1 nmol/L). To rationalize this, the hCAII-(4) adduct, obtained by cocrystallization, reveals not only the sulfamate group and the backbone of (4) interacting with the catalytic site and the associated hydrophobic pocket, respectively, but also the potential H-bonding between the N-(pyridin-3-ylmethyl) group and Nepsilon(2) of Gln(136). Like (2), both (10) and its phenolic precursor (9) are non-estrogenic using a uterine weight gain assay. In summary, a highly potent, long-acting, and nonestrogenic steroid sulfatase inhibitor was designed with hCAII inhibitory properties that should positively influence in vivo behavior. Compound (10) and other related inhibitors of this structural class further expand the armory of steroid sulfatase inhibitors against hormone-dependent breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Fluorine/chemistry , Steryl-Sulfatase/antagonists & inhibitors , Animals , Chromatography, Liquid , Crystallography, X-Ray , Female , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Conformation , Rats , Rats, Wistar , Spectrometry, Mass, Electrospray Ionization , Steryl-Sulfatase/chemistry
14.
J Med Chem ; 51(14): 4226-38, 2008 Jul 24.
Article in English | MEDLINE | ID: mdl-18590272

ABSTRACT

To explore aromatase inhibition and to broaden the structural diversity of dual aromatase-sulfatase inhibitors (DASIs), we introduced the steroid sulfatase (STS) inhibitory pharmacophore to letrozole. Letrozole derivatives were prepared bearing bis-sulfamates or mono-sulfamates with or without adjacent substituents. The most potent of the achiral and racemic aromatase inhibitor was 40 (IC 50 = 3.0 nM). Its phenolic precursor 39 was separated by chiral HPLC, and the absolute configuration of each enantiomer was determined using vibrational and electronic circular dichroism in tandem with calculations of the predicted spectra. Of the two enantiomers, ( R)-phenol ( 39a) was the most potent aromatase inhibitor (IC 50 = 0.6 nM, comparable to letrozole), whereas the ( S)-sulfamate, ( 40b) inhibited STS most potently (IC 50 = 553 nM). These results suggest that a new structural class of DASI for potential treatment of hormone-dependent breast cancer has been identified, and this is the first report of STS inhibition by an enantiopure nonsteroidal compound.


Subject(s)
Aromatase Inhibitors/pharmacology , Aromatase/drug effects , Nitriles/pharmacology , Steryl-Sulfatase/antagonists & inhibitors , Triazoles/pharmacology , Aromatase Inhibitors/chemistry , Chromatography, High Pressure Liquid , Crystallography, X-Ray , Humans , Letrozole , Molecular Structure , Nitriles/chemistry , Stereoisomerism , Structure-Activity Relationship , Triazoles/chemistry
15.
Endocrinology ; 149(8): 4035-42, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18450955

ABSTRACT

The past few years have seen an increase in the reported incidence of endometrial carcinoma, one of the most frequently diagnosed malignancies of the female genital tract. Estrogen production is vital for the mitogenesis of endometrial tumors. Inhibition of steroid sulfatase (STS), an enzyme responsible for the synthesis of steroids with estrogenic properties, may represent a novel therapeutic target for this type of cancer. This study investigates the effects of STX64 (also known as 667Coumate and BN83495) and STX213, two potent STS inhibitors, on hormone-dependent endometrial cancer cell growth in vivo. When tested in intact mice with endometrial cancer xenografts, STX64 had limited effect on tumor growth. In contrast, the microtubule disruptor STX140 reduced tumor growth by 55%. In a hormone-dependent endometrial xenograft model in ovariectomized mice, both STX64 and STX213 given orally, daily at 1 mg/kg significantly inhibited tumor growth by 48 and 67%, respectively. However, when given orally at 1 mg/kg once weekly, only STX213 still inhibited tumor proliferation. At a higher dose of STX64 (10 mg/kg, orally, daily), a greater tumor growth inhibition of 59% was observed. Liver and tumor STS activity was completely inhibited in all daily treatment groups. Plasma estradiol (E2) levels were also significantly decreased. A significant correlation was observed between plasma E2 concentrations and STS activity, indicating the importance of circulating E2 on tumor growth. This novel study demonstrates for the first time that STS inhibitors are potent inhibitors of endometrial cancer growth in nude mice.


Subject(s)
Azasteroids/therapeutic use , Carcinoma/drug therapy , Coumarins/therapeutic use , Endometrial Neoplasms/drug therapy , Steryl-Sulfatase/antagonists & inhibitors , Sulfonamides/therapeutic use , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Carcinoma/pathology , Cell Proliferation/drug effects , Endometrial Neoplasms/pathology , Estradiol/analogs & derivatives , Estradiol/blood , Estradiol/pharmacology , Estrenes/pharmacology , Female , Humans , Mice , Mice, Nude , Models, Biological , Neoplasms, Hormone-Dependent , Ovariectomy , Sulfonic Acids , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
ChemMedChem ; 3(4): 603-18, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18236493

ABSTRACT

The synthesis and in vitro biological evaluation (JEG-3 cells) of a series of novel and potent aromatase inhibitors, prepared by microwave-enhanced Suzuki cross-coupling methodology, are reported. These compounds possess a biphenyl template incorporated with the haem-ligating triazolylmethyl moiety, either on its own or in combination with other substituent(s) at various positions on the phenyl rings. The most potent aromatase inhibitor reported herein has an IC(50) value of 0.12 nM, although seven of its congeners are also highly potent (IC(50)

Subject(s)
Aromatase Inhibitors/chemical synthesis , Biphenyl Compounds/chemistry , Aromatase Inhibitors/chemistry , Aromatase Inhibitors/pharmacology , Cells, Cultured , Humans , Models, Molecular , Structure-Activity Relationship
17.
Chembiochem ; 9(4): 613-23, 2008 Mar 03.
Article in English | MEDLINE | ID: mdl-18288656

ABSTRACT

Pseudomonas aeruginosa arylsulfatase catalyses the cleavage of aryl sulfates and is an excellent model for human estrone sulfatase, which is implicated in hormone-dependent breast cancer. Aryl sulfamates are inactivators of sulfatases; however, little is known about their mechanism. We studied the inactivation of Pseudomonas aeruginosa arylsulfatase A by a range of aryl sulfamates, including the clinical agent 667COUMATE (STX64) used to inactivate estrone sulfatase. Inactivation was time dependent, irreversible, and active-site directed, consistent with a covalent modification at the active site. In terms of the kinetic parameters of inactivation k(inact) and K(i), K(i) values are in the micromolar to nanomolar range, and the inactivation half-life is less than 30 s. A Brønsted plot of k(inact)/K(i) has a steep slope (beta(lg) = -1.1), which implies that the transition state for the first irreversible chemical step of inactivation involves a high degree of charge transfer and cleavage of the ArO-S bond. Detection of the released phenol and titration of the residual activity showed the stoichiometry of inactivation to be in the range 3-6, with the greatest values found for the most effective inactivators. Thus, multiple sulfamoylation events appear to occur during the inactivation process. These data provide valuable insight into the mechanism of sulfatase inactivation by sulfamates.


Subject(s)
Argon/chemistry , Arylsulfatases/metabolism , Oxygen/chemistry , Pseudomonas aeruginosa/enzymology , Sulfonic Acids/chemistry , Sulfonic Acids/metabolism , Sulfur/chemistry , Arylsulfatases/genetics , Binding Sites , Enzyme Activation , Hydrogen-Ion Concentration , Molecular Structure , Phenol/chemistry , Phenol/metabolism , Protein Processing, Post-Translational , Pseudomonas aeruginosa/genetics , Substrate Specificity , Titrimetry
18.
Breast Cancer Res Treat ; 111(1): 129-38, 2008 Sep.
Article in English | MEDLINE | ID: mdl-17914670

ABSTRACT

Estrogenic steroids, such as estradiol, are known to play a crucial role in the development and growth of hormone-dependent breast cancer. Steroid sulfatase (STS) inhibitors that can prevent the biosynthesis of these steroids via the sulfatase pathway offer therapeutic potential. We show here the in vivo profile, including the efficacy in a xenograft breast cancer model and pharmacokinetics, of three potent STS inhibitors. MCF-7 cells stably over-expressing STS cDNA (MCF-7STS) were generated. Ovariectomised, MF-1, female nude mice receiving subcutaneous injections of estradiol sulfate (E2S) and bearing MCF-7STS xenografts, were orally treated with the STS inhibitors STX64, STX213, and STX1938. Treatment was administered once weekly at a dose of 1 mg/kg for 35 days during which animals received E2S thrice weekly. Mice were weighed and tumor measurements taken weekly. Furthermore, the pharmacokinetics for STX213 was determined in rats. STX213 and STX1938 exhibited potent STS inhibition in vivo. However, STX1938 demonstrated a greater duration of activity. In vehicle treated nude mice receiving E2S, tumor volumes increased by 260% after 35 days compared to day zero. STX64 (1 mg/kg) failed to reduce tumor growth when given once weekly. STX213 and STX1938 (once weekly, 1 mg/kg) significantly inhibited (P < 0.05) tumor growth over this same time period. These compounds completely inhibited liver and tumor STS activity and significantly reduced the levels of plasma E2. This study indicates that the STS inhibitor, STX213, exhibits excellent efficacy and pharmacokinetics and therefore offers a potentially novel treatment for hormone-dependent breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Azasteroids/pharmacology , Enzyme Inhibitors/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Steryl-Sulfatase/antagonists & inhibitors , Sulfonic Acids/pharmacology , Animals , Female , Humans , Mice , Mice, Nude , Neoplasms, Hormone-Dependent/drug therapy , Xenograft Model Antitumor Assays
19.
Org Biomol Chem ; 5(20): 2940-52, 2007 Oct 21.
Article in English | MEDLINE | ID: mdl-17972413

ABSTRACT

The synthesis and biological evaluation of a series of novel Dual Aromatase-Sulfatase Inhibitors (DASIs) are described. It is postulated that dual inhibition of the aromatase and steroid sulfatase enzymes, both responsible for the biosynthesis of oestrogens, will be beneficial in the treatment of hormone-dependent breast cancer. The compounds are based upon the Anastrozole aromatase inhibitor template which, while maintaining the haem ligating triazole moiety crucial for enzyme inhibition, was modified to include a phenol sulfamate ester motif, the pharmacophore for potent irreversible steroid sulfatase inhibition. Adaption of a synthetic route to Anastrozole was accomplished via selective radical bromination and substitution reactions to furnish a series of aromatase inhibitory pharmacophores. Linking these fragments to the phenol sulfamate ester moiety employed SN2, Heck and Mitsunobu reactions with phenolic precursors, from where the completed DASIs were achieved via sulfamoylation. In vitro, the lead compound, 11, had a high degree of potency against aromatase (IC50 3.5 nM), comparable with that of Anastrozole (IC50 1.5 nM) whereas, only moderate activity against steroid sulfatase was found. However, in vivo, 11 surprisingly exhibited potent dual inhibition. Compound 11 was modelled into the active site of a homology model of human aromatase and the X-ray crystal structure of steroid sulfatase.


Subject(s)
Aromatase Inhibitors/chemistry , Aromatase/drug effects , Enzyme Inhibitors/chemistry , Nitriles/chemistry , Sulfatases/antagonists & inhibitors , Triazoles/chemistry , Anastrozole , Animals , Aromatase Inhibitors/chemical synthesis , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Humans , Molecular Conformation , Rats , Rats, Wistar , Structure-Activity Relationship
20.
Org Biomol Chem ; 5(18): 2940-52, 2007 Sep 21.
Article in English | MEDLINE | ID: mdl-17728860

ABSTRACT

The synthesis and biological evaluation of a series of novel Dual Aromatase-Sulfatase Inhibitors (DASIs) are described. It is postulated that dual inhibition of the aromatase and steroid sulfatase enzymes, both responsible for the biosynthesis of oestrogens, will be beneficial in the treatment of hormone-dependent breast cancer. The compounds are based upon the Anastrozole aromatase inhibitor template which, while maintaining the haem ligating triazole moiety crucial for enzyme inhibition, was modified to include a phenol sulfamate ester motif, the pharmacophore for potent irreversible steroid sulfatase inhibition. Adaption of a synthetic route to Anastrozole was accomplished via selective radical bromination and substitution reactions to furnish a series of inhibitory aromatase pharmacophores. Linking these fragments to the phenol sulfamate ester moiety employed S(N)2, Heck and Mitsunobu reactions with phenolic precursors, from where the completed DASIs were achieved via sulfamoylation. In vitro, the lead compound, 11, had a high degree of potency against aromatase (IC(50) 3.5 nM), comparable with that of Anastrozole (IC(50) 1.5 nM) whereas, only moderate activity against steroid sulfatase was found. However, in vivo, 11 surprisingly exhibited potent dual inhibition. Compound 11 was modelled into the active site of a homology model of human aromatase and the X-ray crystal structure of steroid sulfatase.


Subject(s)
Aromatase Inhibitors/chemical synthesis , Aromatase Inhibitors/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Nitriles/chemistry , Sulfatases/antagonists & inhibitors , Triazoles/chemistry , Anastrozole , Aromatase Inhibitors/chemistry , Enzyme Inhibitors/chemistry , Magnetic Resonance Spectroscopy , Models, Molecular , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...