Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Pharmacol Res ; 128: 52-60, 2018 02.
Article in English | MEDLINE | ID: mdl-29287690

ABSTRACT

The aminosteroid derivative RM-133 is an effective anticancer molecule for which proof of concept has been achieved in several mouse xenograph models (HL-60, MCF-7, PANC-1 and OVCAR-3). To promote this new family of molecules toward a clinical phase 1 trial, the mechanism of action governing the anticancer properties of the representative candidate RM-133 needs to be characterized. In vitro experiments were first used to determine that RM-133 causes apoptosis in cancer cells. Then, using proteomic and transcriptomic experiments, RM-133 cytotoxicity was proven to be achieved via the endoplasmic reticulum (ER)-related apoptosis, which characterizes RM-133 as an endoplasmic reticulum stress aggravator (ERSA) anticancer drug. Furthermore, an shRNA-genome-wide screening has permitted to identify the steroidogenic acute regulator-related lipid transfer protein 5 (STARD5) as a major player in the RM-133 ER-related apoptosis mechanism, which was validated by an in vitro binding experiment. Altogether, the results presented herein suggest that RM-133 provokes a disturbance of cholesterol homeostasis via the implication of STARD5, which delivers an ERSA molecule to the ER. These results will be a springboard for RM-133 in its path toward clinical use.


Subject(s)
Androstenes/pharmacology , Antineoplastic Agents/pharmacology , Carrier Proteins/metabolism , Cholesterol/metabolism , Endoplasmic Reticulum Stress , Adaptor Proteins, Vesicular Transport , Apoptosis/drug effects , Carrier Proteins/genetics , Cell Line, Tumor , Homeostasis/drug effects , Humans
2.
PLoS One ; 12(2): e0171871, 2017.
Article in English | MEDLINE | ID: mdl-28182747

ABSTRACT

In the fight against androgen-sensitive prostate cancer, the enzyme 17ß-hydroxysteroid dehydrogenase type 3 (17ß-HSD3) is an attractive therapeutic target considering its key role in the formation of androgenic steroids. In this study, we attempted to assess the in vivo efficacy of the compound RM-532-105, an androsterone derivative developed as an inhibitor of 17ß-HSD3, in the prostate cancer model of androgen-sensitive LAPC-4 cells xenografted in nude mice. RM-532-105 did not inhibit the tumor growth induced by 4-androstene-3,17-dione (4-dione); rather, the levels of the androgens testosterone (T) and dihydrotestosterone (DHT) increased within the tumors. In plasma, however, DHT levels increased but T levels did not. In troubleshooting experiments, the non-androgenic potential of RM-532-105 was confirmed by two different assays (LAPC-4 proliferation and androgen receptor transcriptional activity assays). The enzyme 5α-reductase was also revealed to be the predominant enzyme metabolizing 4-dione in LAPC-4 cells, yielding 5α-androstane-3,17-dione and not T. Other 17ß-HSDs than 17ß-HSD3 seem responsible in the androgen synthesis. From experiments with LAPC-4 cells, we fortuitously came across the interesting finding that 17ß-HSD3 inhibitor RM-532-105 is concentrated inside tumors.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Androstanes/therapeutic use , Enzyme Inhibitors/therapeutic use , Prostatic Neoplasms/drug therapy , Sulfonamides/therapeutic use , Androstanes/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Dihydrotestosterone/blood , Enzyme Inhibitors/pharmacology , Humans , Male , Mice , Mice, Nude , Sulfonamides/pharmacology
3.
Bioorg Med Chem Lett ; 26(9): 2179-83, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27025340

ABSTRACT

The steroidogenic enzyme 17ß-hydroxysteroid dehydrogenase type 3 (17ß-HSD3) is a therapeutic target in the management of androgen-sensitive diseases such as prostate cancer and benign prostate hyperplasia. In this Letter, we designed and synthesized the first fluorescent inhibitor of this enzyme by combining a fluorogenic dansyl moiety to the chemical structure of a known inhibitor of 17ß-HSD3. The synthesized compound 3 is a potent fluorogenic compound (λex=348 nm and λ em=498 nm). It crosses the cell membrane, keeps its fluorescent properties and is distributed inside the LNCaP cells overexpressing 17ß-HSD3, where it inhibits the transformation of 4-androstene-3,17-dione into the androgen testosterone (IC50=262 nM).


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Androstanes/pharmacology , Dansyl Compounds/pharmacology , Fluorescent Dyes/pharmacology , Androstanes/chemical synthesis , Cell Line, Tumor , Cell Membrane/metabolism , Dansyl Compounds/chemical synthesis , Flow Cytometry , Fluorescent Dyes/chemical synthesis , Humans , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology
4.
Steroids ; 107: 55-64, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26742630

ABSTRACT

Steroids possessing an ethynyl group at position 17α (tertiary alcohols) are well known to be more stable than their non-ethynyl analogs (secondary alcohols). To facilitate the development of new drugs with better metabolic stability, we developed a new diethylsilyl acetylenic linker allowing us to rapidly synthesize libraries of ethynylated steroid derivatives using a solid-phase strategy. To illustrate its usefulness, this linker was used to expand the molecular diversity of a lead compound having a hydroxy acetylenic pattern and to potentially find new compounds with interesting cytotoxic activity against leukemia cell lines. Herein, we report the chemical synthesis and the characterization of three libraries of ethynylated aminosteroid derivatives using the diethylacetylenic linker. We discuss their antiproliferative activities obtained in 2 leukemia cell lines (HL-60 and Jurkat), which results provided new structure-activity relationships. We also identified a new promising aminosteroid derivative with an azetidine moiety (compound B1) inhibiting 60% and 75% of HL-60 and Jurkat cell proliferation, respectively, at 1 µM. More generally, these results validate the use of a diethylsilyl acetylenic linker for researchers interested in generating libraries of alcohol derivatives with better stability and drug profile.


Subject(s)
Amines , Antineoplastic Agents , Cell Proliferation/drug effects , Leukemia/drug therapy , Steroids , Amines/chemical synthesis , Amines/chemistry , Amines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , HL-60 Cells , Humans , Jurkat Cells , Leukemia/metabolism , Leukemia/pathology , Steroids/chemical synthesis , Steroids/chemistry , Steroids/pharmacology
5.
PLoS One ; 10(12): e0144890, 2015.
Article in English | MEDLINE | ID: mdl-26660672

ABSTRACT

Ovarian and pancreatic cancers are two of the most aggressive and lethal cancers, whose management faces only limited therapeutic options. Typically, these tumors spread insidiously accompanied first with atypical symptoms, and usually shift to a drug resistance phenotype with the current pharmaceutical armamentarium. Thus, the development of new drugs acting via a different mechanism of action represents a clear priority. Herein, we are reporting for the first time that the aminosteroid derivative RM-133, developed in our laboratory, displays promising activity on two models of aggressive cancers, namely ovarian (OVCAR-3) and pancreatic (PANC-1) cancers. The IC50 value of RM-133 was 0.8 µM and 0.3 µM for OVCAR-3 and PANC-1 cell lines in culture, respectively. Based on pharmacokinetic studies on RM-133 using 11 different vehicles, we selected two main vehicles: aqueous 0.4% methylcellulose:ethanol (92:8) and sunflower oil:ethanol (92:8) for in vivo studies. Using subcutaneous injection of RM-133 with the methylcellulose-based vehicle, growth of PANC-1 tumors xenografted to nude mice was inhibited by 63%. Quite interestingly, RM-133 injected subcutaneously with the methylcellulose-based or sunflower-based vehicles reduced OVCAR-3 xenograft growth by 122% and 100%, respectively. After the end of RM-133 treatment using the methylcellulose-based vehicle, OVCAR-3 tumor growth inhibition was maintained for ≥ 1 week. RM-133 was also well tolerated in the whole animal, no apparent sign of toxicity having been detected in the xenograft studies.


Subject(s)
Androstenes/therapeutic use , Antineoplastic Agents/therapeutic use , Ovarian Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Androstenes/blood , Androstenes/toxicity , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/pathology , Pancreatic Neoplasms/pathology , Transplantation, Heterologous
6.
Bioorg Med Chem ; 23(17): 5433-51, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26277760

ABSTRACT

17ß-Hydroxysteroid dehydrogenase type 3 (17ß-HSD3) is a key enzyme involved in the biosynthesis of testosterone and dihydrotestosterone. These hormones are known to stimulate androgen-dependent prostate cancer. In order to generate effective inhibitors of androgen biosynthesis without androgenic effect, we synthesized a new family of 3-spiromorpholinone and 3-spirocarbamate androsterone derivatives bearing diversified hydrophobic groups. We also tested their inhibitory activity in a microsomal fraction of 17ß-HSD3-containing rat testes, and their androgenic effect on androgen-sensitive LAPC-4 cells. From our first structure-activity relationship (SAR) study, we noted that compound 7e inhibited 17ß-HSD3 (77% at 0.1 µM) compared to our reference compound RM-532-105 (76% at 0.1 µM), but exhibited a residual androgenic effect. A library of 7e analogue compounds was next synthesized in order to generate compounds with reduced androgenic activity. In this new SAR study, the sulfonamide compound 7e21 and the carboxamide compound 7e22 inhibited 17ß-HSD3 (IC50 = 28 and 88 nM, respectively). These two compounds were not androgenic and not cytotoxic even at the highest concentration tested, but their inhibitory activity decreased in intact LNCaP cells overexpressing 17ß-HSD3 (LNCaP[17ß-HSD3]). Structural modifications of these two lead compounds could however be tested to produce a second generation of 17ß-HSD3 inhibitors.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Carbamates/chemistry , Carbamates/pharmacology , Morpholines/chemistry , Morpholines/pharmacology , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , 17-Hydroxysteroid Dehydrogenases/metabolism , Androgens/chemistry , Androgens/pharmacology , Animals , Cell Line, Tumor , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Male , Prostate/drug effects , Prostate/enzymology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Rats
7.
J Steroid Biochem Mol Biol ; 153: 170-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26291835

ABSTRACT

17ß-Hydroxysteroid dehydrogenase type 3 (17ß-HSD3 or HSD17B3) catalyzes the last step in the biosynthesis of the potent androgen testosterone (T), by stereoselectively reducing the C17 ketone of 4-androstene-3,17-dione (4-dione), with NADPH as cofactor. Since T plays an important role in androgen-sensitive diseases, this enzyme is thus an interesting therapeutic target. In an attempt to design compounds to lower the level of T, we synthesized androsterone derivatives substituted at position 3 as inhibitors of 17ß-HSD3, and selected one of the most potent compounds for additional studies. In an enzymatic assay in homogenized and whole HEK-293 cells overexpressing 17ß-HSD3, the inhibitor RM-532-105 efficiently inhibited the conversion of natural substrate 4-dione (50nM) into T with an IC50 of 26nM and 5nM, respectively. Moreover, the inhibitor RM-532-105 (10mg/kg) reached a plasma concentration of 250ng/mL at 7h (AUC 24h: 3485ngh/mL) after subcutaneous (s.c.) injection in the rat. In order to mimic the human situation in which 4-dione is converted to T in the testis, we used intact rats. Treatment for 7 days with 17ß-HSD3 inhibitor RM-532-105 by s.c. injection or oral gavage exerted no effect on the testis, prostate and seminal vesicle weight and no modification in the levels of plasma steroids. However, after this treatment, the concentration of inhibitor in plasma increased depending on the dose. We thereafter determined the concentration of inhibitor in the testis and we discovered that the compound was slightly present. In fact, at 10mg/kg, the inhibitor RM-532-105 seems to have difficulty penetrating inside the testis and was found to be concentrated in the testicular capsule, and therefore unable to inhibit the 17ß-HSD3 located inside the testis. However, with a higher dose of 50mg/kg injected s.c. in rats, RM-532-105 significantly decreased the level of T and dihydrotestosterone measured in plasma at 2h.

8.
J Steroid Biochem Mol Biol ; 141: 44-51, 2014 May.
Article in English | MEDLINE | ID: mdl-24434282

ABSTRACT

17ß-Hydroxysteroid dehydrogenase type 3 (17ß-HSD3 or HSD17B3) catalyzes the last step in the biosynthesis of the potent androgen testosterone (T), by stereoselectively reducing the C17 ketone of 4-androstene-3,17-dione (4-dione), with NADPH as cofactor. Since T plays an important role in androgen-sensitive diseases, this enzyme is thus an interesting therapeutic target. In an attempt to design compounds to lower the level of T, we synthesized androsterone derivatives substituted at position 3 as inhibitors of 17ß-HSD3, and selected one of the most potent compounds for additional studies. In an enzymatic assay in homogenized and whole HEK-293 cells overexpressing 17ß-HSD3, the inhibitor RM-532-105 efficiently inhibited the conversion of natural substrate 4-dione (50nM) into T with an IC50 of 26nM and 5nM, respectively. Moreover, the inhibitor RM-532-105 (10mg/kg) reached a plasma concentration of 250ng/mL at 7h (AUC 24h: 3485ngh/mL) after subcutaneous (s.c.) injection in the rat. In order to mimic the human situation in which 4-dione is converted to T in the testis, we used intact rats. Treatment for 7 days with 17ß-HSD3 inhibitor RM-532-105 by s.c. injection or oral gavage exerted no effect on the testis, prostate and seminal vesicle weight and no modification in the levels of plasma steroids. However, after this treatment, the concentration of inhibitor in plasma increased depending on the dose. We thereafter determined the concentration of inhibitor in the testis and we discovered that the compound was slightly present. In fact, at 10mg/kg, the inhibitor RM-532-105 seems to have difficulty penetrating inside the testis and was found to be concentrated in the testicular capsule, and therefore unable to inhibit the 17ß-HSD3 located inside the testis. However, with a higher dose of 50mg/kg injected s.c. in rats, RM-532-105 significantly decreased the level of T and dihydrotestosterone measured in plasma at 2h.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Androstanes/pharmacology , Sulfonamides/pharmacology , Testosterone/blood , 17-Hydroxysteroid Dehydrogenases/metabolism , Androstanes/pharmacokinetics , Androstenedione/blood , Animals , Dihydrotestosterone/blood , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Inhibitory Concentration 50 , Luteinizing Hormone/blood , Male , Prostate/drug effects , Prostate/metabolism , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacokinetics , Testis/drug effects , Testis/enzymology
9.
Bioorg Med Chem Lett ; 23(23): 6360-2, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24144853

ABSTRACT

Spiromorpholinone derivatives were synthesized from androsterone or cyclohexanone in 6 or 3 steps, respectively, and these scaffolds were used for the introduction of a hydrophobic group via a nucleophilic substitution. Non-steroidal spiromorpholinones are not active as inhibitors of 17ß-hydroxysteroid dehydrogenase type 3 (17ß-HSD3), but steroidal morpholinones are very potent inhibitors. In fact, those with (S) stereochemistry are more active than their (R) homologues, whereas N-benzylated compounds are more active than their non substituted precursors. The target compounds exhibited strong inhibition of 17ß-HSD3 in rat testis homogenate (87-92% inhibition at 1 µM).


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Androsterone/analogs & derivatives , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , 17-Hydroxysteroid Dehydrogenases/metabolism , Androsterone/chemical synthesis , Animals , Male , Rats , Structure-Activity Relationship , Testis/enzymology
10.
Acta Crystallogr C ; 68(Pt 6): o231-4, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22669194

ABSTRACT

The title compounds, (3R,5S,5'R,8R,9S,10S,13S,14S)-10,13-dimethyl-5'-(2-methylpropyl)tetradecahydro-6'H-spiro[cyclopenta[a]phenanthrene-3,2'-[1,4]oxazinane]-6',17(2H)-dione, C(26)H(41)NO(3), (I), and methyl (2R)-2-[(3R,5S,8R,9S,10S,13S,14S)-10,13-dimethyl-2',17-dioxohexadecahydro-3'H-spiro[cyclopenta[a]phenanthrene-3,5'-[1,3]oxazolidin-3'-yl]]-4-methylpentanoate, C(28)H(43)NO(5), (II), possess the typical steroid shape (A-D rings), but they differ in their extra E ring. The azalactone E ring in (I) shows a half-chair conformation, while the carbamate E ring of (II) is planar. The orientation of the E-ring substituent is clearly established and allows a rationalization of the biological results obtained with such androsterone derivatives.


Subject(s)
Androgen Antagonists/chemistry , Androgen Antagonists/pharmacology , Androgens/biosynthesis , Androgens/chemistry , Androsterone/chemistry , Androsterone/pharmacology , Androsterone/analogs & derivatives , Crystallography, X-Ray , Models, Molecular
SELECTION OF CITATIONS
SEARCH DETAIL
...