Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters










Publication year range
1.
J Steroid Biochem Mol Biol ; 242: 106544, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38754521

ABSTRACT

Sex steroid hormones such as estrogen estradiol (E2) and androgen dihydrotestosterone (DHT) are involved in the development of hormone-dependent cancers. Blockade of 17ß-hydroxysteroid dehydrogenase type 7 (17ß-HSD7), a member of the short chain dehydrogenase/reductase superfamily, is thought to decrease E2 levels while increasing those of DHT. Therefore, its unique double action makes this enzyme as an interesting drug target for treatment of breast cancer. The chemical synthesis, molecular characterization, and preliminary biological evaluation as 17ß-HSD7 inhibitors of novel carbamate derivatives 3 and 4 are described. Like previous 17ß-HSD7 inhibitors 1 and 2, compounds 3 and 4 bear a hydrophobic nonyl side chain at the C-17ß position of a 4-aza-5α-androstane nucleus, but compound 3 has an oxygen atom replacing the CH2 in the steroid A-ring C-2 position, while compound 4 has a C17-spiranic E-ring containing a carbamate function. They both inhibited the in vitro transformation of estrone (E1) into E2 by 17ß-HSD7, but the introduction of a (17 R)-spirocarbamate is preferable to replacing C-2 methylene with an oxygen atom since compound 4 (IC50 = 63 nM) is an inhibitor 14 times more powerful than compound 3 (IC50 = 900 nM). Furthermore, when compared to the reference inhibitor 1 (IC50 = 111 nM), the use of a C17-spiranic E-ring made it possible to introduce differently the hydrophobic nonyl side chain, without reducing the inhibitory activity.


Subject(s)
17-Hydroxysteroid Dehydrogenases , Enzyme Inhibitors , 17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/metabolism , Humans , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Estradiol/chemistry , Estradiol/metabolism , Estradiol/pharmacology , Carbamates/chemistry , Carbamates/pharmacology , Carbamates/chemical synthesis , Estrone/chemistry , Estrone/pharmacology , Estrone/chemical synthesis
2.
Antioxidants (Basel) ; 13(4)2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38671833

ABSTRACT

In osteoarthritis (OA), oxidative stress plays a crucial role in maintaining and sustaining cartilage degradation. Current OA management requires a combination of pharmaceutical and non-pharmacological strategies, including intraarticular injections of hyaluronic acid (HA). However, several lines of evidence reported that HA oxidation by reactive oxygen species (ROS) is linked with HA cleavage and fragmentation, resulting in reduced HA viscosity. Resolvin D1 (RvD1) is a lipid mediator that is biosynthesized from omega-3 polyunsaturated fatty acids and is a good candidate with the potential to regulate a panoply of biological processes, including tissue repair, inflammation, oxidative stress, and cell death in OA. Herein, newly designed and synthesized imidazole-derived RvD1 analogues were introduced to compare their potential antioxidant properties with commercially available RvD1. Their antioxidant capacities were investigated by several in vitro chemical assays including oxygen radical absorbance capacity, 2,2-diphenyl-1-picrylhydrazyl radical scavenging, ferric ion reducing antioxidant power, hydroxyl radical scavenging, and HA fragmentation assay. All results proved that imidazole-derived RvD1 analogues showed excellent antioxidant performance compared to RvD1 due to their structural modifications. Interestingly, they scavenged the formed reactive oxygen species (ROS) and protected HA from degradation, as verified by agarose gel electrophoresis and gel permission chromatography. A computational study using Gaussian 09 with DFT calculations and a B3LYP/6-31 G (d, p) basis set was also employed to study the relationship between the antioxidant properties and chemical structures as well as calculation of the molecular structures, frontier orbital energy, molecular electrostatic potential, and bond length. The results showed that the antioxidant activity of our analogues was higher than that of RvD1. In conclusion, the findings suggest that imidazole-derived RvD1 analogues can be good candidates as antioxidant molecules for the treatment of oxidative stress-related diseases like OA. Therefore, they can prolong the longevity of HA in the knee and thus may improve the mobility of the articulation.

3.
J Med Virol ; 96(3): e29484, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38402600

ABSTRACT

Antiviral therapy based on neuraminidase (oseltamivir) or polymerase (baloxavir marboxil) inhibitors plays an important role in the management of influenza infections. However, the emergence of drug resistance and the uncontrolled inflammatory response are major limitations in the treatment of severe influenza disease. Protectins D1 (PD1) and DX (PDX), part of a family of pro-resolving mediators, have previously demonstrated anti-influenza activity as well as anti-inflammatory properties in various clinical contexts. Herein, we synthetized a series of simplified PDX analogs and assessed their in vitro antiviral activity against influenza A(H1N1) viruses, including oseltamivir- and baloxavir-resistant variants. In ST6GalI-MDCK cells, the PDX analog AN-137B reduced viral replication in a dose-dependent manner with IC50 values of 23.8 for A/Puerto Rico/8/1934 (H1N1) and between 32.6 and 36.7 µM for susceptible and resistant A(H1N1)pdm09 viruses. In MTS-based cell viability experiments, AN-137B showed a 50% cellular cytotoxicity (CC50 ) of 638.7 µM with a resulting selectivity index of 26.8. Of greater importance, the combination of AN-137B with oseltamivir or baloxavir resulted in synergistic and additive in vitro effects, respectively. Treatment of lipopolysaccharide (LPS)-stimulated macrophages with AN-137B resulted in a decrease of iNOS activity as shown by the reduction of nitrite production, suggesting an anti-inflammatory effect. In conclusion, our results indicate that the protectin analog AN-137B constitutes an interesting therapeutic modality against influenza A virus, warranting further evaluation in animal models.


Subject(s)
Dibenzothiepins , Docosahexaenoic Acids , Influenza A Virus, H1N1 Subtype , Influenza A virus , Influenza, Human , Morpholines , Pyridones , Triazines , Animals , Humans , Oseltamivir/pharmacology , Oseltamivir/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Influenza, Human/drug therapy , Anti-Inflammatory Agents/therapeutic use , Drug Resistance, Viral , Neuraminidase
4.
J Clin Med ; 12(13)2023 Jun 24.
Article in English | MEDLINE | ID: mdl-37445276

ABSTRACT

Breast cancer (BC) is a heterogenous disease classified into four molecular subtypes (Luminal A, Luminal B, HER2 and triple-negative (TNBC)) depending on the expression of the estrogen receptor (ER), the progesterone receptor (PR) and the human epidermal receptor 2 (HER2). The development of effective treatments for BC, especially TNBC, remains a challenge. Aminosteroid derivative RM-581 has previously shown an antiproliferative effect in multiple cancers in vitro and in vivo. In this study, we evaluated its effect in BC cell lines representative of BC molecular subtypes, including metastatic TNBC. We found that RM-581 has an antiproliferative effect on all BC molecular subtypes, especially on Luminal A and TNBC, in 2D and 3D cultures. The combination of RM-581 and trastuzumab or trastuzumab-emtansine enhanced the anticancer effect of each drug for HER2-positive BC cell lines, and the combination of RM-581 and taxanes (docetaxel or paclitaxel) improved the antiproliferative effect of RM-581 in TNBC and metastatic TNBC cell lines. We also confirmed that RM-581 is an endoplasmic reticulum (EnR)-stress aggravator by inducing an increase in EnR-stress-induced apoptosis markers such as BIP/GRP78 and CHOP and disrupting lipid homeostasis. This study demonstrates that RM-581 could be effective for the treatment of BC, especially TNBC.

5.
Cancers (Basel) ; 15(11)2023 Jun 02.
Article in English | MEDLINE | ID: mdl-37296995

ABSTRACT

The aminosteroid derivative RM-581 blocks with high potency the growth of androgen-dependent (AR+) prostate cancer VCaP, 22Rv1, and LAPC-4 cells. Notably, RM-581 demonstrated superior antiproliferative activity in LAPC-4 cells compared to enzalutamide and abiraterone, two drugs that exhibited a synergistic effect in combination with RM-581. These findings suggest that RM-581 may have an action that is not directly associated with the hormonal pathway of androgens. Furthermore, RM-581 completely blocks tumor growth in LAPC-4 xenografts when given orally at 3, 10, and 30 mg/kg in non-castrated (intact) nude mice. During this study, an accumulation of RM-581 was observed in tumors compared to plasma (3.3-10 folds). Additionally, the level of fatty acids (FA) increased in the tumors and livers of mice treated with RM-581 but not in plasma. The increase was greater in unsaturated FA (21-28%) than in saturated FA (7-11%). The most affected FA were saturated palmitic acid (+16%), monounsaturated oleic acid (+34%), and di-unsaturated linoleic acid (+56%), i.e., the 3 most abundant FA, with a total of 55% of the 56 FA measured. For cholesterol levels, there was no significant difference in the tumor, liver, or plasma of mice treated or not with RM-581. Another important result was the innocuity of RM-581 in mice during a 28-day xenograft experiment and a 7-week dose-escalation study, suggesting a favorable safety window for this new promising drug candidate when given orally.

6.
J Org Chem ; 88(11): 7088-7095, 2023 Jun 02.
Article in English | MEDLINE | ID: mdl-37172290

ABSTRACT

We report a gram-scale total synthesis of protectin DX (PDX) following a convergent synthetic route (24 steps) from l-malic acid. This novel synthetic strategy is based on the assembly of three main building blocks using a Sonogashira coupling reaction (blocks A and B) and Wittig olefination (block C) to provide the 22-carbon backbone of PDX. A key stereoselective reduction of enediyne leads to a central E,Z,E-trienic system of PDX and also gives access to its labeled versions (D and T).

7.
Molecules ; 28(2)2023 Jan 07.
Article in English | MEDLINE | ID: mdl-36677690

ABSTRACT

Estradiol (E2) plays an important role in the progression of diseases such as breast cancer and endometriosis. Inhibition of 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD1), the enzyme that catalyzes the last step in the biosynthesis of the estrogenic hormone E2, therefore constitutes an interesting approach for the treatment of these two estrogen-dependent diseases. In order to obtain new inhibitors of 17ß-HSD1, the impact of a m-carbamoylphenyloxy group at position three of an estrane nucleus was evaluated by preparing three derivatives of estrone (E1) and E2 using a microwave-assisted synthesis of diaryl ethers. Their inhibitory activity was addressed on two cell lines (T-47D and Z-12) representative of breast cancer and endometriosis, respectively, but unlike T-47D cells, Z-12 cells were not found suitable for testing potential 17ß-HSD1 inhibitors. Thus, the addition of the m-carbamoylphenyl group at C3 of E1 (compound 5) did not increase the inhibition of E1 to E2 transformation by 17ß-HSD1 present in T-47D cells (IC50 = 0.31 and 0.21 µM for 5 and E1, respectively), and this negative effect was more obvious for E2 derivatives 6 and 10 (IC50 = 1.2 and 1.3 µM, respectively). Molecular docking allowed us to identify key interactions with 17ß-HSD1 and to highlight these new inhibitors' actions through an opposite orientation than natural enzyme substrate E1's classical one. Furthermore, molecular modeling experiments explain the better inhibitory activity of E1-ether derivative 5, as opposed to the E2-ether derivatives 6 and 10. Finally, when tested on T-47D and Z-12 cells, compounds 5, 6 and 10 did not stimulate the proliferation of these two estrogen-dependent cell lines. In fact, they reduced it.


Subject(s)
17-Hydroxysteroid Dehydrogenases , Breast Neoplasms , Endometriosis , Enzyme Inhibitors , Female , Humans , 17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Estradiol , Estrogens , Estrone/pharmacology , Molecular Docking Simulation
8.
Bioorg Chem ; 129: 106145, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36174444

ABSTRACT

Prostate cancer is the most common cancer among men and the development of new therapeutic agents is needed for its treatment and/or diagnosis. 17ß-hydroxysteroid dehydrogenase type 3 (17ß-HSD3) is involved in the production of androgens, which stimulates the proliferation of prostate cancer cells. Piperazinomethyl-androsterone sulfonamide derivatives were developed as 17ß-HSD3 inhibitors and the concentration of a representative sulfonamide derivative (compound 1) was found to accumulate in prostate tumor tissues relatively to plasma in a mouse xenograft experiment. This finding gives us the opportunity to specifically target the prostate cancer tumors through the development of a radiolabelled version of compound 1 toward targeted molecular radiotherapy or radioimaging diagnosis. The chemical synthesis of fluorinated and iodinated analogs of compound 1 was achieved, leading to a series of compounds with similar levels of inhibition as the initial candidate. From 17ß-HSD3 inhibition activity, molecular modeling and mouse plasma-concentration studies, the most promising compound of this series was selected, its 18F-radiolabelled version (18F-3) synthesized, and imaging/biodistribution studies engaged. When injected in mice, however, 18F-3 uptake in the target tissues (LNCaP[17ß-HSD3] tumors and testicles) was not sufficient to allow their visualization by positron emission tomography. Plasma concentration values of compounds 3-8 administered orally, however, showed that the para-iodo compound 7 is the most metabolically stable and could therefore be an interesting alternative for radiolabelling and radiotreatment.


Subject(s)
Enzyme Inhibitors , Prostatic Neoplasms , Male , Humans , Mice , Animals , Tissue Distribution , Enzyme Inhibitors/chemistry , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Sulfonamides/pharmacology
9.
J Steroid Biochem Mol Biol ; 222: 106136, 2022 09.
Article in English | MEDLINE | ID: mdl-35691460

ABSTRACT

Endometriosis is a gynecological disorder affecting about 10% of women and can lead to invalidating painful symptoms and infertility. Since there is no current definitive cure for this disease, new therapeutic options are necessary. 17ß-Hydroxysteroid dehydrogenase type 1 (17ß-HSD1) is involved in the production of estradiol (E2), the most potent estrogen in women, and of 5-androstene-3ß,17ß-diol (5-diol), a weaker estrogen than E2, but whose importance increases after menopause. 17ß-HSD1 is therefore a pharmacological target of choice for the treatment of estrogen-dependent diseases such as endometriosis. We developed a targeted-covalent (irreversible) and non-estrogenic inhibitor of 17ß-HSD1, a molecule named PBRM, and herein evaluated its efficiency for the treatment of endometriosis. In a cell-free assay containing estrone (E1), the natural substrate of 17ß-HSD1, PBRM was able to block the formation of E2 in a collection of 50 human endometriosis lesions from a different clinical feature type, location, and phase. When given orally by gavage at 15 mg/kg to baboons, the resulting plasmatic concentration of PBRM was found to be sufficiently high (up to 125 ng/mL) for an efficacy study in a non-human primate (baboon) endometriosis model. After 2 months of treatment, the number of lesions/adhesions decreased in 60% of animals (3/5) in the PBRM-treated group, compared to the placebo group which showed an increase in the number of lesion/adhesions in 60% (3/5) of animals. Indeed, the total number of lesions/adhesions decreased in treated group (-6.5 or -19% when excluding one animal) while it increased in the control group receiving a placebo (+11%). Analysis of specific endometriotic lesions revealed that PBRM decreased the number of red lesions (-67%; 8/12) and white lesions (-35%; 11/31), but not of blue-black lesions. Similarly, PBRM decreased the surface area of dense adhesions and filmy adhesions, as compared to placebo. Also, PBRM treatment did not significantly affect the number of menstrual days. Finally, this targeted covalent inhibitor showed no adverse effects and no apparent toxicity for the duration of the treatment. These data indicate that 17ß-HSD1 inhibitor PBRM is a promising candidate for therapy targeting endometriosis and supports the need of additional efforts toward clinical trials.


Subject(s)
Endometriosis , Estradiol , 17-Hydroxysteroid Dehydrogenases , Animals , Endometriosis/drug therapy , Enzyme Inhibitors/pharmacology , Estradiol/chemistry , Estradiol/pharmacology , Estradiol Dehydrogenases , Estrogens , Female , Humans , Primates
10.
Curr Org Synth ; 19(7): 838-845, 2022.
Article in English | MEDLINE | ID: mdl-35473530

ABSTRACT

AIMS AND OBJECTIVE: The syntheses of glucuronide metabolites of phenolic xenoestrogens triclosan and 2-phenylphenol, namely triclosan-O-glucuronide (TCS-G; 1), and 2-phenylphenol-Oglucuronide (OPP-G; 2), were achieved for use as analytical standards. METHODS: Under classical conditions previously reported for glucuronide synthesis, the final basic hydrolysis of the peracylated ester intermediate leading to the free glucuronides is often a limiting step. Indeed, the presence of contaminating by-products resulting from ester elimination has often been observed during this step. This is particularly relevant when the sugar unit is close to a crowded environment as for triclosan and 2-phenylphenol. RESULTS: To circumvent these problems, we proposed mild conditions for the deprotection of peracetylated glucuronate intermediates. CONCLUSION: A new methodology using a key imidate following a two-step protocol for acetates and methyl ester hydrolysis was successfully applied to the preparation of TCS-d3 (1) and OPP-G (2) as well as deuterated isotopomers TCS-d3-G (1-d3) and OPP-d5-G (2-d5).


Subject(s)
Glucuronides , Triclosan , Esters , Glucuronides/metabolism , Phenols , Reference Standards , Triclosan/metabolism
11.
Int J Mol Sci ; 22(20)2021 Oct 17.
Article in English | MEDLINE | ID: mdl-34681843

ABSTRACT

Aminosteroid derivative RM-581 was previously identified as an endoplasmic-reticulum (ER) stress inducer with potent in vitro and in vivo anticancer activities. We report its evaluation in androgen-independent prostate cancer (PC-3) cells. RM-581 efficiently blocks PC-3 cell proliferation with stronger activity than that of a selection of known antineoplastic agents. This later also showed a synergistic effect with docetaxel, able to block the proliferation of docetaxel-resistant PC-3 cells and, contrary to docetaxel, did not induce cell resistance. RM-581 induced an increase in the expression level of ER stress-related markers of apoptosis, potentially triggered by the presence of RM-581 in the ER of PC-3 cells. These in vitro results were then successfully translated in vivo in a PC-3 xenograft tumor model in nude mice, showing superior blockade than that of docetaxel. RM-581 was also able to stop the progression of PC-3 cells when they had become resistant to docetaxel treatment. Concomitantly, we observed a decrease in gene markers of mevalonate and fatty acid pathways, and intratumoral levels of cholesterol by 19% and fatty acids by 22%. Overall, this work demonstrates the potential of an ER stress inducer as an anticancer agent for the treatment of prostate cancers that are refractory to commonly used chemotherapy treatments.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Endoplasmic Reticulum Stress , Estranes/pharmacology , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation , Docetaxel/therapeutic use , Estranes/therapeutic use , Humans , Male , Mice , Mice, Nude , PC-3 Cells , Prostatic Neoplasms/physiopathology , Xenograft Model Antitumor Assays
12.
ACS Med Chem Lett ; 12(5): 822-826, 2021 May 13.
Article in English | MEDLINE | ID: mdl-34055232

ABSTRACT

RM-581 is an aminosteroid derivative comprised of a steroid core and a quinoline side chain showing potent cytotoxic activity on several types of cancer cells but for which the mechanism of action (MoA) remains to be fully elucidated. The opportunity to turn RM-581 into a fluorescent probe was explored because the addition of a N-dimethyl group was recently reported to induce fluorescence to quinoline derivatives. After the chemical synthesis of the N-dimethyl analogue of RM-581 (RM-581-Fluo), its fluorescent properties, as well as its cytotoxic activity in breast cancer MCF-7 cells, were confirmed. A cell imaging experiment in MCF-7 cells using confocal microscopy then revealed that RM-581-Fluo accumulated into the endoplasmic reticulum (ER) as highlighted by its colocalization with an ER-Tracker dye. This work provides a new tool for RM-581 MoA investigations as well as being a relevant example of a tailor-made quinolone-fluorescent version of a bioactive molecule.

13.
Steroids ; 172: 108856, 2021 08.
Article in English | MEDLINE | ID: mdl-33945801

ABSTRACT

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.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Breast Neoplasms/drug therapy , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Steryl-Sulfatase/antagonists & inhibitors , Trientine/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Structure-Activity Relationship , Tumor Cells, Cultured
14.
Cancers (Basel) ; 13(8)2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33924352

ABSTRACT

17ß-Hydroxysteroid dehydrogenase type 1 (17ß-HSD1) plays an important role in estrogen-dependent breast tumor growth. In addition to being involved in the production of estradiol (E2), the most potent estrogen in women, 17ß-HSD1 is also responsible for the production of 5-androsten-3ß,17ß-diol (5-diol), a weaker estrogen than E2, but whose importance increases after menopause. 17ß-HSD1 is therefore a target of choice for the treatment of estrogen-dependent diseases such as breast cancer and endometriosis. After we developed the first targeted-covalent (irreversible) and non-estrogenic inhibitor of 17ß-HSD1, a molecule named PBRM, our goal was to demonstrate its therapeutic potential. Enzymatic assays demonstrated that estrone (E1) and dehydroepiandrosterone (DHEA) were transformed into E2 and 5-diol in T-47D human breast cancer cells, and that PBRM was able to block these transformations. Thereafter, we tested PBRM in a mouse tumor model (cell-derived T-47D xenografts). After treatment of ovariectomized (OVX) mice receiving E1 or DHEA, PBRM given orally was able to reduce the tumor growth at the control (OVX) level without any observed toxic effects. Thanks to its irreversible type of inhibition, PBRM retained its anti-tumor growth effect, even after reducing its frequency of administration to only once a week, a clear advantage over reversible inhibitors.

15.
J Steroid Biochem Mol Biol ; 210: 105846, 2021 06.
Article in English | MEDLINE | ID: mdl-33609690

ABSTRACT

A new androsterone derivative bearing a 16ß-picolyl group (compound 5; FCO-586-119) was synthetized in four steps from the lead compound 1 (RM-532-105). We measured its inhibitory activity on 17ß-HSD3 using microsomal fraction of rat testes as well as transfected LNCaP[17ß-HSD3] cells. We then assessed its metabolic stability as well as its cytotoxic effect against a panel of cancer cell lines. The addition of a picolyl moiety at C-16 of RM-532-105 steroid core improves the 17ß-HSD3 inhibitory activity in the microsomal fraction of rat testes, but not in whole LNCaP[17ß-HSD3] cells. Interestingly, this structural modification enhances 3-fold the metabolic stability in conjunction with a significant cytotoxic effect against pancreatic, ovarian, breast, lung, and prostate cancer cells. Because the inhibitory activity data against 17ß-HSD3 suggested that both steroid derivatives are non-competitive inhibitors, we performed docking and molecular dynamics simulations using a homology model of this membrane-associated enzyme. The results of these simulations revealed that both RM-532-105 (1) and FCO-586-119 (5) can compete for the cofactor-binding site displaying better binding energy than NADP+.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Androsterone/chemistry , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/metabolism , Androstanes/chemistry , Androsterone/analogs & derivatives , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Stability , Enzyme Inhibitors/chemical synthesis , Humans , Magnetic Resonance Spectroscopy , Male , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Rats, Sprague-Dawley , Sulfonamides/chemistry
16.
Eur J Med Chem ; 209: 112909, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33081987

ABSTRACT

17beta-Hydroxysteroid dehydrogenase type 10 (17ß-HSD10) is the only mitochondrial member of 17ß-HSD family. This enzyme can oxidize estradiol (E2) into estrone (E1), thus reducing concentration of this neuroprotective steroid. Since 17ß-HSD10 possesses properties that suggest a possible role in Alzheimer's disease, its inhibition appears to be a therapeutic strategy. After we identified the androsterone (ADT) derivative 1 as a first steroidal inhibitor of 17ß-HSD10, new analogs were synthesized to increase the metabolic stability, to improve the selectivity of inhibition over 17ß-HSD3 and to optimize the inhibitory potency. From six D-ring derivatives of 1 (17-CO), two compounds (17ß-H/17α-OH and 17ß-OH/17α-CCH) were more metabolically stable and did not inhibit the 17ß-HSD3. Moreover, solid phase synthesis was used to extend the molecular diversity on the 3ß-piperazinylmethyl group of the steroid base core. Eight over 120 new derivatives were more potent inhibitors than 1 for the transformation of E2 to E1, with the 4-(4-trifluoromethyl-3-methoxybenzyl)piperazin-1-ylmethyl-ADT (D-3,7) being 16 times more potent (IC50 = 0.14 µM). Finally, D-ring modification of D-3,7 provided 17ß-OH/17α-CCH derivative 25 and 17ß-H/17α-OH derivative 26, which were more potent inhibitor than 1 (1.8 and 2.4 times, respectively).


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Alzheimer Disease/drug therapy , Enzyme Inhibitors/chemical synthesis , Small Molecule Libraries/chemistry , Steroids/chemical synthesis , Biocatalysis , Drug Evaluation, Preclinical , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Estradiol/chemistry , Estrone/chemistry , HEK293 Cells , Humans , Piperazine/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Solid-Phase Synthesis Techniques , Steroids/metabolism , Steroids/pharmacology , Structure-Activity Relationship
17.
Bioorg Med Chem ; 28(7): 115368, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32122754

ABSTRACT

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.


Subject(s)
Estradiol/analogs & derivatives , Steryl-Sulfatase/antagonists & inhibitors , Estradiol/chemical synthesis , Estrone/chemistry , HEK293 Cells , Humans , Microscopy, Confocal , Models, Molecular , Molecular Structure , Optical Imaging , Quantitative Structure-Activity Relationship
18.
Eur J Med Chem ; 188: 111990, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-31893547

ABSTRACT

The aminosteroid (AM) RM-581 is built around a mestranol backbone and has recently emerged as this family's lead candidate, showing in vitro and in vivo potency over different types of cancer, including high fatality pancreatic cancer. To extend the structure-activity relationships (SAR) to other estrane analogs, we synthesized a focused series of RM-581 derivatives at position C3 or C2 of its steroidal core. These new AM derivatives were first tested on a large selection of prostate, breast, pancreatic and ovarian cancer cell lines. The impact of these modifications on metabolic stability (human liver microsomes) was also measured. A SAR study revealed a fine regulation of anticancer activity related to the nature of the substituent. Indeed, the addition of potential prodrug groups like acetate, sulfamate or phosphate (compounds 8, 9 and 10) at C3 of the phenolic counterpart provided better antiproliferative activities than RM-581 in breast and pancreatic cancer cell types while maintaining activity in other cancer cell lines. Also, the phosphate group was highly beneficial on water solubility. However, the bulkier carbamate prodrugs 6 (N,N-dimethyl) and 7 (N,N-diethyl) were less active. Otherwise, carbon homologation (CH2) at C2 (compound 33) was beneficial to metabolic stability and, in the meantime, this AM conserved the same anticancer activity as RM-581. However, the replacement of the hydroxy or methoxy at C3 by a hydrogen or an acetyl (compound 17 or 21b) was detrimental for anticancer activity, pointing to a crucial molecular interaction of the aromatic oxygen atom at this position. Overall, this work provided a better knowledge of the structural requirements to maintain RM-581's anticancer activity, and also identified minor structural modifications to increase both metabolic stability and water solubility, three important parameters of pharmacological development.


Subject(s)
Antineoplastic Agents/pharmacology , Estranes/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Estranes/chemistry , Estranes/metabolism , Humans , Liver/chemistry , Liver/metabolism , Molecular Structure , Solubility , Structure-Activity Relationship , Tumor Cells, Cultured , Water/chemistry
19.
Bioorg Med Chem Lett ; 30(2): 126783, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31753699

ABSTRACT

5α-Dihydrotestosterone (5α-DHT) possesses a great affinity for the androgen receptor (AR), and its binding to AR promotes the proliferation of prostate cancer (PC) cells in androgen-dependent PC. Primarily synthesized from testosterone (T) in testis, 5α-DHT could also be produced from 5α-androstane-3α,17ß-diol (3α-diol), an almost inactive androgen, following non-classical pathways. We reported the chemical synthesis of non-commercially available [4-14C]-3α-diol from [4-14C]-T, and the development of a biological assay to identify inhibitors of the 5α-DHT formation from radiolabeled 3α-diol in LAPC-4 cell PC model. We measured the inhibitory potency of 5α-androstane derivatives against the formation of 5α-DHT, and inhibition curves were obtained for the most potent compounds (IC50 = 1.2-14.1 µM). The most potent inhibitor 25 (IC50 = 1.2 µM) possesses a 4-(4-CF3-3-CH3O-benzyl)piperazinyl methyl side chain at C3ß and 17ß-OH/17α-CCH functionalities at C17 of a 5α-androstane core.


Subject(s)
Androgens/metabolism , Androstane-3,17-diol/metabolism , Dihydrotestosterone/metabolism , Androstane-3,17-diol/chemistry , Cell Line, Tumor , Dihydrotestosterone/chemistry , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Structure-Activity Relationship
20.
J Med Chem ; 62(15): 7070-7088, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31268309

ABSTRACT

Decreasing the intratumoral androgen biosynthesis by using an inhibitor of 17ß-hydroxysteroid dehydrogenase type 3 (17ß-HSD3) is a strategy to treat prostate cancer. The androsterone (ADT) derivative 1 (RM-532-105) has shown strong inhibitory activity on 17ß-HSD3, but needs to be improved. Herein, we describe the chemical synthesis and characterization of two series of analogues to address the impact of A- and D-ring modifications on 17ß-HSD3 inhibitory activity, androgenic effect, and metabolic stability. Structure-activity relationships were generated by adding different groups at C16/C17 (D-ring diversification) or replacing the ADT backbone by a nor-androstane or an estrane backbone (A-ring diversification). D-ring derivatives were less potent inhibitors than lead compound 1, whereas steroidal backbone (A-ring) change led to identifying promising novel estrane derivatives. This culminated with potent 17ß-HSD3 inhibitors 23, 27, 31, and 33 (IC50 = 0.10, 0.02, 0.13, and 0.17 µM, respectively), which did not stimulate LAPC-4 cell proliferation and displayed higher plasma concentration in mice than lead compound 1.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/chemistry , Androsterone/analogs & derivatives , Androsterone/pharmacology , Androsterone/therapeutic use , Animals , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Mice , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...