1
|
Poirier D. Recent advances in the development of 17beta-hydroxysteroid dehydrogenase inhibitors. Steroids 2025; 213:109529. [PMID: 39532224 DOI: 10.1016/j.steroids.2024.109529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
The family of 17β-hydroxysteroid dehydrogenases (17β-HSDs) occupies a prominent place due to its number of isoforms, which carry out a bidirectional transformation (reduction of a steroid carbonyl to alcohol and oxidation of a steroid alcohol to ketone) depending on the nature of the cofactor present. Involved in the activation or inactivation of key estrogens and androgens, 17β-HSDs are therefore therapeutic targets whose selective inhibition would make it possible to be considered for the treatment of several diseases, such as breast cancer, prostate cancer, endometriosis, Alzheimer's disease and osteoporosis. This review article is a continuation of those having reported the great diversity of inhibitors developed over the last years but focusses on inhibitors recently developed. Work to obtain more effective inhibitors that target the first known isoforms (types 1, 2, 3, 5 and 7) has continued, among others, but new inhibitors that target the isoforms more recently reported in the literature (types 10, 12, 13 and 14) are now being reported. Dual inhibitors of two enzymes (17β-HSD1 and steroid sulfatase) were also reported. These inhibitors were grouped according to the 17β-HSD type inhibited and their backbone (steroidal or non-steroidal) when necessary. They were also reported in chronological order and according to the research group.
Collapse
Affiliation(s)
- Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec Research Center-Université Laval, Québec, QC G1V 4G2, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
2
|
He J, Ji Z, Sang J, Quan H, Zhang H, Lu H, Zheng J, Wang S, Ge RS, Li X. Potent inhibition of human and rat 17β-hydroxysteroid dehydrogenase 1 by curcuminoids and the metabolites: 3D QSAR and in silico docking analysis. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:433-456. [PMID: 38785078 DOI: 10.1080/1062936x.2024.2355529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024]
Abstract
Curcumin, an extensively utilized natural pigment in the food industry, has attracted considerable attention due to its potential therapeutic effects, such as anti-tumorigenic and anti-inflammatory activities. The enzyme 17β-Hydroxysteroid dehydrogenase 1 (17β-HSD1) holds a crucial position in oestradiol production and exhibits significant involvement in oestrogen-responsive breast cancers and endometriosis. This study investigated the inhibitory effects of curcuminoids, metabolites, and analogues on 17β-HSD1, a key enzyme in oestradiol synthesis. Screening 10 compounds, including demethoxycurcumin (IC50, 3.97 μM) and dihydrocurcumin (IC50, 5.84 μM), against human and rat 17β-HSD1 revealed varying inhibitory potencies. These compounds suppressed oestradiol secretion in human BeWo cells at ≥ 5-10 μM. 3D-Quantitative structure-activity relationship (3D-QSAR) and molecular docking analyses elucidated the interaction mechanisms. Docking studies and Gromacs simulations suggested competitive or mixed binding to the steroid or NADPH/steroid binding sites of 17β-HSD1. Predictive 3D-QSAR models highlighted the importance of hydrophobic regions and hydrogen bonding in inhibiting 17β-HSD1 activity. In conclusion, this study provides valuable insights into the inhibitory effects and mode of action of curcuminoids, metabolites, and analogues on 17β-HSD1, which may have implications in the field of hormone-related disorders.
Collapse
Affiliation(s)
- J He
- Department of Anesthesiology, Yuying Children's Hospital, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| | - Z Ji
- Department of Anesthesiology, Yuying Children's Hospital, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| | - J Sang
- Department of Anesthesiology, Yuying Children's Hospital, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| | - H Quan
- Department of Anesthesiology, Yuying Children's Hospital, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| | - H Zhang
- Department of Anesthesiology, Yuying Children's Hospital, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| | - H Lu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - J Zheng
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - S Wang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - R S Ge
- Department of Anesthesiology, Yuying Children's Hospital, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, Wenzhou, Zhejiang Province, China
| | - X Li
- Department of Anesthesiology, Yuying Children's Hospital, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, Wenzhou, Zhejiang Province, China
| |
Collapse
|
3
|
Kuzminac IZ, Bekić SS, Ćelić AS, Jakimov DS, Sakač MN. Antitumor potential of novel 5α,6β-dibromo steroidal D-homo lactone. Steroids 2022; 188:109118. [PMID: 36183814 DOI: 10.1016/j.steroids.2022.109118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 01/11/2023]
Abstract
New steroidal D-homo androstane derivative, 5α,6β-dibromo-3β-hydroxy-17-oxa-17a-homoandrostan-16-one was synthesized and its structure was confirmed by NMR spectroscopy. In silico ADME properties of this compound were assessed using the SwissADME online prediction tool. Six human cancer cell lines (MDA-MB-231, MCF-7, PC3, HT-29, HeLa, and A549) and one human noncancerous cell line (MRC-5) were used for in vitro cytotoxicity testing. Novel steroidal dibromide was also tested for relative binding affinity for the ligand binding domain of estrogen receptor α and β or the androgen receptor using a published assay in yeast cells. Ligand binding domains of each steroid receptor were expressed in-frame with yellow fluorescent protein in yeast and the fluorescence intensity changes upon addition of test compound was measured. The new compound showed selective cytotoxic activity against HT-29 (colon adenocarcinoma) and A549 (lung adenocarcinoma) cell lines, as well as the potential to induce apoptosis in HT-29 cells, while results obtained from ligand binding assay in yeast suggested a lack of significant estrogenic or androgenic properties.
Collapse
Affiliation(s)
- Ivana Z Kuzminac
- University of Novi Sad, Faculty of Sciences, Department of Chemistry, Biochemistry and Environmental Protection, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia.
| | - Sofija S Bekić
- University of Novi Sad, Faculty of Sciences, Department of Chemistry, Biochemistry and Environmental Protection, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia
| | - Anđelka S Ćelić
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia
| | - Dimitar S Jakimov
- Oncology Institute of Vojvodina, Faculty of Medicine, University of Novi Sad, Put Dr Goldmana 4, 21204 Sremska Kamenica, Serbia
| | - Marija N Sakač
- University of Novi Sad, Faculty of Sciences, Department of Chemistry, Biochemistry and Environmental Protection, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia
| |
Collapse
|
4
|
Mercorio A, Giampaolino P, Romano A, Dällenbach P, Pluchino N. Is intracrinology of endometriosis relevant in clinical practice? A systematic review on estrogen metabolism. Front Endocrinol (Lausanne) 2022; 13:950866. [PMID: 36204107 PMCID: PMC9531311 DOI: 10.3389/fendo.2022.950866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
UNLABELLED Endometriosis is a chronic, multifactorial, estrogen-dependent disease. The abnormal endocrine microenvironment of endometriosis lesions is considered a main feature and multiple enzymatic pathways leading to local increased synthesis of estrogens have been identified. However, the relevance of intracrinology in clinical practice is still lacking. Medline, Embase, Scopus database were systematically searched for studies reporting on local estrogens metabolism of endometriotic lesions. The main enzymatic pathways involved in the intracrinology of endometriosis such as aromatase (CYP19A1), 17β-hydroxysteroid dehydrogenase (HSD17B) type 1, type 2 and type 5, steroid sulfatase (STS), estrogen sulfotransferase (SULT1E1) were assessed with a critical perspective on their role in disease endocrine phenotyping, drug resistance and as therapeutic targets. Overall, studies heterogeneity and missing clinical data affect the interpretation of the clinical role of these enzymes. Although the use of some drugs such as aromatase inhibitors has been proposed in clinical practice for two decades, their potential clinical value is still under investigation as well as their modality of administration. A closer look at new, more realistic drug targets is provided and discussed. Altered expression of these key enzymes in the lesions have far reaching implication in the development of new drugs aimed at decreasing local estrogenic activity with a minimal effect on gonadal function; however, given the complexity of the evaluation of the expression of the enzymes, multiple aspects still remains to be clarified. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022311329, identifier CRD42022311329.
Collapse
Affiliation(s)
- Antonio Mercorio
- Department of Pediatrics Gynecology and Obstetrics, Division of Gynecology, Geneva University Hospitals, Geneva, Switzerland
| | - Pierluigi Giampaolino
- Department of Public Health, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Andrea Romano
- Obstetrics and Gynaecology Department, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
| | - Patrick Dällenbach
- Department of Pediatrics Gynecology and Obstetrics, Division of Gynecology, Geneva University Hospitals, Geneva, Switzerland
| | - Nicola Pluchino
- Department of Pediatrics Gynecology and Obstetrics, Division of Gynecology, Geneva University Hospitals, Geneva, Switzerland
- *Correspondence: Nicola Pluchino,
| |
Collapse
|
5
|
Herman BE, Gardi J, Julesz J, Tömböly C, Szánti-Pintér E, Fehér K, Skoda-Földes R, Szécsi M. Steroidal ferrocenes as potential enzyme inhibitors of the estrogen biosynthesis. Biol Futur 2021; 71:249-264. [PMID: 34554507 DOI: 10.1007/s42977-020-00023-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 06/04/2020] [Indexed: 01/13/2023]
Abstract
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 17β-hydroxysteroid dehydrogenase type 1 (17β-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 17β-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 17β-estradiol in target tissues.
Collapse
Affiliation(s)
- Bianka Edina Herman
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, P. O. Box 427, Szeged, 6720, Hungary
| | - János Gardi
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, P. O. Box 427, Szeged, 6720, Hungary
| | - János Julesz
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, P. O. Box 427, Szeged, 6720, Hungary
| | - Csaba Tömböly
- Laboratory of Chemical Biology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Temesvári körút 62, P. O. Box 521, Szeged, 6726, Hungary
| | - Eszter Szánti-Pintér
- Department of Organic Chemistry, Institute of Chemistry, University of Pannonia, Egyetem utca 10, P. O. Box 158, Veszprém, 8200, Hungary
| | - Klaudia Fehér
- Department of Organic Chemistry, Institute of Chemistry, University of Pannonia, Egyetem utca 10, P. O. Box 158, Veszprém, 8200, Hungary
| | - Rita Skoda-Földes
- Department of Organic Chemistry, Institute of Chemistry, University of Pannonia, Egyetem utca 10, P. O. Box 158, Veszprém, 8200, Hungary.
| | - Mihály Szécsi
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, P. O. Box 427, Szeged, 6720, Hungary.
| |
Collapse
|
6
|
Ferrante T, Adinolfi S, D'Arrigo G, Poirier D, Daga M, Lolli ML, Balliano G, Spyrakis F, Oliaro-Bosso S. Multiple catalytic activities of human 17β-hydroxysteroid dehydrogenase type 7 respond differently to inhibitors. Biochimie 2019; 170:106-117. [PMID: 31887335 DOI: 10.1016/j.biochi.2019.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 12/26/2019] [Indexed: 10/25/2022]
Abstract
Cholesterol biosynthesis is a multistep process in mammals that includes the aerobic removal of three methyl groups from the intermediate lanosterol, one from position 14 and two from position 4. During the demethylations at position 4, a 3-ketosteroid reductase catalyses the conversion of both 4-methylzymosterone and zymosterone to 4-methylzymosterol and zymosterol, respectively, restoring the alcoholic function of lanosterol, which is also maintained in cholesterol. Unlike other eukaryotes, mammals also use the same enzyme as an estrone reductase that can transform estrone (E1) into estradiol (E2). This enzyme, named 17β-hydroxysteroid dehydrogenase type 7 (HSD17B7), is therefore a multifunctional protein in mammals, and one that belongs to both the HSD17B family, which is involved in steroid-hormone metabolism, and to the family of post-squalene cholesterol biosynthesis enzymes. In the present study, a series of known inhibitors of human HSD17B7's E1-reductase activity have been assayed for potential inhibition against 3-ketosteroid reductase activity. Surprisingly, the assayed compounds lost their inhibition activity when tested in HepG2 cells that were incubated with radiolabelled acetate and against the recombinant overexpressed human enzyme incubated with 4-methylzymosterone (both radiolabelled and not). Preliminary kinetic analyses suggest a mixed or non-competitive inhibition on the E1-reductase activity, which is in agreement with Molecular Dynamics simulations. These results raise questions about the mechanism(s) of action of these possible inhibitors, the enzyme dynamic regulation and the interplay between the two activities.
Collapse
Affiliation(s)
- Terenzio Ferrante
- Department of Science and Drug Technology, University of Torino, Via P. Giuria 9, 10125, Turin, Italy
| | - Salvatore Adinolfi
- Department of Science and Drug Technology, University of Torino, Via P. Giuria 9, 10125, Turin, Italy
| | - Giulia D'Arrigo
- Department of Science and Drug Technology, University of Torino, Via P. Giuria 9, 10125, Turin, Italy
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, CHU de Québec - Research Centre and Université Laval, 2705, Boulevard Laurier T-4-50 Québec, G1V 4G2, Canada
| | - Martina Daga
- Department of Science and Drug Technology, University of Torino, Via P. Giuria 9, 10125, Turin, Italy
| | - Marco Lucio Lolli
- Department of Science and Drug Technology, University of Torino, Via P. Giuria 9, 10125, Turin, Italy
| | - Gianni Balliano
- Department of Science and Drug Technology, University of Torino, Via P. Giuria 9, 10125, Turin, Italy
| | - Francesca Spyrakis
- Department of Science and Drug Technology, University of Torino, Via P. Giuria 9, 10125, Turin, Italy
| | - Simonetta Oliaro-Bosso
- Department of Science and Drug Technology, University of Torino, Via P. Giuria 9, 10125, Turin, Italy.
| |
Collapse
|
7
|
Maltais R, Poirier D. Development of a Gram-Scale Synthesis of PBRM, an Irreversible Inhibitor of 17β-Hydroxysteroid Dehydrogenase Type 1. Org Process Res Dev 2019. [DOI: 10.1021/acs.oprd.8b00402] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- René Maltais
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec—Research Center (CHUL, T4-42), 2705 Laurier Boulevard, Québec, Québec G1V 4G2, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec—Research Center (CHUL, T4-42), 2705 Laurier Boulevard, Québec, Québec G1V 4G2, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Québec G1V 0A6, Canada
| |
Collapse
|
8
|
Lespérance M, Barbeau X, Roy J, Maltais R, Lagüe P, Poirier D. Chemical synthesis of C3-oxiranyl/oxiranylmethyl-estrane derivatives targeted by molecular modeling and tested as potential inhibitors of 17β-hydroxysteroid dehydrogenase type 1. Steroids 2018; 140:104-113. [PMID: 30273695 DOI: 10.1016/j.steroids.2018.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 11/23/2022]
Abstract
17β-Hydroxysteroid dehydrogenase type 1 (17β-HSD1) is a promising therapeutic target known to play a pivotal role in the progression of estrogen-dependent diseases such as breast cancer, and endometriosis. This enzyme is responsible for the last step in the biosynthesis of the most potent estrogen, estradiol (E2) and its inhibition would prevent the growth of estrogen-sensitive tumors. Based on molecular modeling with docking experiments, we identified two promising C3-oxiranyl/oxiranylmethyl-estrane derivatives that would bind competitively and irreversibly in the catalytic site of 17β-HSD1. They have been synthesized in a short and efficient route and their inhibitory activities over 17β-HSD1 have been assessed by an enzymatic assay. Compound 15, with an oxiranylmethyl group at position C3, was more likely to bind the catalytic site and showed an interesting, but weak, inhibitory activity with an IC50 value of 1.3 µM (for the reduction of estrone into E2 in T-47D cells). Compound 11, with an oxiranyl at position C3, produced a lower inhibition rate, and the IC50 value cannot be determined. When tested in estrogen-sensitive T-47D cells, both compounds were also slightly estrogenic, although much less than the estrogenic hormone E2.
Collapse
Affiliation(s)
- Maxime Lespérance
- Laboratory of Medicinal Chemistry, Oncology and Nephrology Unit, CHU de Québec - Research Center (CHUL T4), Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Xavier Barbeau
- Department of Chemistry, Faculty of Science and Engineering, Université Laval, Québec, QC, Canada; Institut de Biologie Intégrative et des Systèmes (IBIS), and Centre de Recherche sur la Fonction, la Structure et l'Ingénierie des Protéines (PROTEO), Université Laval, Québec, QC, Canada
| | - Jenny Roy
- Laboratory of Medicinal Chemistry, Oncology and Nephrology Unit, CHU de Québec - Research Center (CHUL T4), Québec, QC, Canada
| | - René Maltais
- Laboratory of Medicinal Chemistry, Oncology and Nephrology Unit, CHU de Québec - Research Center (CHUL T4), Québec, QC, Canada
| | - Patrick Lagüe
- Department of Biochemistry, Microbiology and Bioinformatic, Faculty of Science and Engineering, Université Laval, Québec, QC, Canada; Institut de Biologie Intégrative et des Systèmes (IBIS), and Centre de Recherche sur la Fonction, la Structure et l'Ingénierie des Protéines (PROTEO), Université Laval, Québec, QC, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, Oncology and Nephrology Unit, CHU de Québec - Research Center (CHUL T4), Québec, QC, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
9
|
Morsy A, Trippier PC. Amyloid-Binding Alcohol Dehydrogenase (ABAD) Inhibitors for the Treatment of Alzheimer’s Disease. J Med Chem 2018; 62:4252-4264. [DOI: 10.1021/acs.jmedchem.8b01530] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Ahmed Morsy
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Paul C. Trippier
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
- Center for Chemical Biology, Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| |
Collapse
|
10
|
Dutour R, Roy J, Cortés-Benítez F, Maltais R, Poirier D. Targeting Cytochrome P450 (CYP) 1B1 Enzyme with Four Series of A-Ring Substituted Estrane Derivatives: Design, Synthesis, Inhibitory Activity, and Selectivity. J Med Chem 2018; 61:9229-9245. [DOI: 10.1021/acs.jmedchem.8b00907] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Raphaël Dutour
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec—Research Center, 2705 Laurier Boulevard, Québec, Québec G1V 4G2, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Québec G1V 4G2, Canada
| | - Jenny Roy
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec—Research Center, 2705 Laurier Boulevard, Québec, Québec G1V 4G2, Canada
| | - Francisco Cortés-Benítez
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec—Research Center, 2705 Laurier Boulevard, Québec, Québec G1V 4G2, Canada
- Department of Pharmacy, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City, 04510, Mexico
| | - René Maltais
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec—Research Center, 2705 Laurier Boulevard, Québec, Québec G1V 4G2, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec—Research Center, 2705 Laurier Boulevard, Québec, Québec G1V 4G2, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Québec G1V 4G2, Canada
| |
Collapse
|
11
|
Collin LJ, Cronin-Fenton DP, Ahern TP, Christiansen PM, Damkier P, Ejlertsen B, Hamilton-Dutoit S, Kjærsgaard A, Silliman RA, Sørensen HT, Lash TL. Cohort Profile: the Predictors of Breast Cancer Recurrence (ProBe CaRE) Premenopausal Breast Cancer Cohort Study in Denmark. BMJ Open 2018; 8:e021805. [PMID: 30068618 PMCID: PMC6074634 DOI: 10.1136/bmjopen-2018-021805] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
PURPOSE The Predictors of Breast Cancer Recurrence (ProBe CaRe) study was established to evaluate modification of tamoxifen (TAM) effectiveness in premenopausal women through reduced activity of TAM-metabolising enzymes. It comprehensively evaluates the effects of pharmacogenetic variants, use of concomitant medications and biomarkers involved in oestrogen metabolism on breast cancer recurrence risk. PARTICIPANTS The ProBe CaRe study was established using resources from the Danish Breast Cancer Group (DBCG), including 5959 premenopausal women diagnosed with stage I-III primary breast cancer between 2002 and 2010 in Denmark. Eligible participants were divided into two groups based on oestrogen receptor alpha (ERα) expression and receipt of TAM therapy, 4600 are classified as ERα+/TAM+ and 1359 are classified as ERα-/TAM-. The ProBe CaRe study is a population-based cohort study nested in a nearly complete source population, clinical, tumour and demographic data were abstracted from DBCG registry data. Linkage to Danish registries allows for abstraction of information regarding comorbid conditions, comedication use and mortality. Formalin-fixed paraffin-embedded tissue samples have been prepared for DNA extraction and immunohistochemical assay. FINDINGS TO DATE To mitigate incorrect classification of patients into specific categories, we conducted a validation substudy. We compared data acquired from registry and from medical record review to calculate positive predictive values (PPVs) and negative predictive values. We observed PPVs near 100% for tumour size, lymph node involvement, receptor status, surgery type, receipt of radiotherapy, receipt of chemotherapy and TAM treatment. We found that the PPVs were 96% (95% CI 83% to 100%) for change in endocrine therapy and 61% (95% CI 42% to 77%) for menopausal transition. FUTURE PLANS The ProBeCaRe cohort study is well positioned to comprehensively examine pharmacogenetic variants. We will use a Bayesian pathway analysis to evaluate the complete TAM metabolic path to allow for gene-gene interactions, incorporating information of other important patient characteristics.
Collapse
Affiliation(s)
- Lindsay J Collin
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | | | - Thomas P Ahern
- Department of Surgery, The Robert Larner, M.D. College of Medicine at The University of Vermont, Burlington, Vermont, USA
| | - Peer M Christiansen
- Breast Unit, Surgical Department, Randers Regional Hospital, Randers, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
- Danish Breast Cancer Group, Copenhagen, Denmark
| | - Per Damkier
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Bent Ejlertsen
- Danish Breast Cancer Group, Copenhagen, Denmark
- Rigshospitalet, Copenhagen, Denmark
| | | | - Anders Kjærsgaard
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Rebecca A Silliman
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
- Boston University School of Medicine, Boston University, Boston, Massachusetts, USA
| | - Henrik Toft Sørensen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Health Research & Policy (Epidemiology), Stanford University, Stanford, California, USA
| | - Timothy L Lash
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
12
|
Li T, Zhu D, Labrie F, Lin S. Crystal Structures of Human 17<i>β</i>-Hydroxysteroid Dehydrogenase Type 1 Complexed with the Dual-Site Inhibitor EM-139. Health (London) 2018. [DOI: 10.4236/health.2018.108081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
13
|
Dutour R, Cortés-Benítez F, Roy J, Poirier D. Structure-Based Design and Synthesis of New Estrane-Pyridine Derivatives as Cytochrome P450 (CYP) 1B1 Inhibitors. ACS Med Chem Lett 2017; 8:1159-1164. [PMID: 29152048 DOI: 10.1021/acsmedchemlett.7b00265] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/11/2017] [Indexed: 12/31/2022] Open
Abstract
Inhibition of cytochrome P450 (CYP) 1B1 is a promising therapeutic strategy, as such an inhibitor could modulate the bioactivation of procarcinogens while reducing drug resistance. Based on docking studies, the synthesis of 12 estra-1,3,5(10)-triene derivatives containing a pyridin-3-/4-yl moiety at position C2, C3, or C4 was performed, and we measured their inhibitory activity on CYP1B1 using the ethoxyresorufin-O-deethylase (EROD) assay. The position of the nitrogen atom in the aromatic ring has little influence on their inhibition potency, but compounds with a pyridinyl at C2 of the steroid nucleus are more potent CYP1B1 inhibitors than those with a pyridinyl at C3 or C4. Estradiol derivatives (OH at C17β) are also 10-fold more potent inhibitors than estrone derivatives (carbonyl at C17). Thus, 2-(pyridin-3-yl)-estradiol (4a) is the best CYP1B1 inhibitor (IC50 = 0.011 μM) from this series of compounds, and the best steroid inhibitor reported until now. It is also 7.5-fold more potent than the well-known nonsteroidal CYP1B1 inhibitor α-naphthoflavone (IC50 = 0.083 μM).
Collapse
Affiliation(s)
- Raphaël Dutour
- Laboratory
of Medicinal Chemistry, Endocrinology-Nephrology Unit, CHU de Québec−Research Center, Québec, Qc, Canada
- Department
of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Qc, Canada
| | - Francisco Cortés-Benítez
- Laboratory
of Medicinal Chemistry, Endocrinology-Nephrology Unit, CHU de Québec−Research Center, Québec, Qc, Canada
- Department
of Pharmacy, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City, Mexico
| | - Jenny Roy
- Laboratory
of Medicinal Chemistry, Endocrinology-Nephrology Unit, CHU de Québec−Research Center, Québec, Qc, Canada
| | - Donald Poirier
- Laboratory
of Medicinal Chemistry, Endocrinology-Nephrology Unit, CHU de Québec−Research Center, Québec, Qc, Canada
- Department
of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, Qc, Canada
| |
Collapse
|
14
|
Insight into the mode of action and selectivity of PBRM, a covalent steroidal inhibitor of 17β-hydroxysteroid dehydrogenase type 1. Biochem Pharmacol 2017; 144:149-161. [DOI: 10.1016/j.bcp.2017.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 08/04/2017] [Indexed: 12/13/2022]
|
15
|
Perreault M, Maltais R, Roy J, Dutour R, Poirier D. Design of a Mestranol 2-N-Piperazino-Substituted Derivative Showing Potent and Selective in vitro and in vivo Activities in MCF-7 Breast Cancer Models. ChemMedChem 2017; 12:177-182. [PMID: 28060448 DOI: 10.1002/cmdc.201600482] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/21/2016] [Indexed: 11/10/2022]
Abstract
Anticancer structure-activity relationship studies on aminosteroid (5α-androstane) derivatives have emerged with a promising lead candidate: RM-133 (2β-[1-(quinoline-2-carbonyl)pyrrolidine-2-carbonyl]-N-piperazine-5α-androstane-3α,17β-diol), which possesses high in vitro and in vivo activities against several cancer cells, and selectivity over normal cells. However, the relatively weak metabolic stability of RM-133 has been a drawback to its progression toward clinical trials. We investigated the replacement of the androstane backbone by a more stable mestranol moiety. The resulting compound, called RM-581 ({4-[17α-ethynyl-17β-hydroxy-3-methoxyestra-1,3,5(10)-trien-2-yl]piperazin-1-yl}[(2S)-1-(quinolin-2-ylcarbonyl)pyrrolidin-2-yl]methanone), was synthesized efficiently in only five steps from commercially available estrone. In comparison with RM-133, RM-581 was found to be twice as metabolically stable, retains potent cytotoxic activity in breast cancer MCF-7 cell culture, and fully blocks tumor growth in a mouse xenograft model of breast cancer. Advantageously, the selectivity over normal cells has been increased with this estrane version of RM-133. In fact, RM-581 showed a better selectivity index (15.3 vs. 3.0) for breast cancer MCF-7 cells over normal breast MCF-10A cells, and was found to be nontoxic toward primary human kidney proximal tubule cells at doses reaching 50 μm.
Collapse
Affiliation(s)
- Martin Perreault
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec-Research Center (CHUL, T4-42), 2705 Laurier Boulevard, Québec City, Québec, G1V 4G2, Canada
| | - René Maltais
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec-Research Center (CHUL, T4-42), 2705 Laurier Boulevard, Québec City, Québec, G1V 4G2, Canada
| | - Jenny Roy
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec-Research Center (CHUL, T4-42), 2705 Laurier Boulevard, Québec City, Québec, G1V 4G2, Canada
| | - Raphaël Dutour
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec-Research Center (CHUL, T4-42), 2705 Laurier Boulevard, Québec City, Québec, G1V 4G2, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec-Research Center (CHUL, T4-42), 2705 Laurier Boulevard, Québec City, Québec, G1V 4G2, Canada.,Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec City, Québec, Canada
| |
Collapse
|
16
|
Targeting cytochrome P450 (CYP) 1B1 with steroid derivatives. Bioorg Med Chem Lett 2016; 26:5272-5276. [PMID: 27687674 DOI: 10.1016/j.bmcl.2016.09.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/15/2016] [Accepted: 09/16/2016] [Indexed: 11/20/2022]
Abstract
Inhibition of cytochrome P450 1B1 (CYP1B1) represents a promising therapeutic strategy, because it would enable action at three different levels: (1) by inhibiting the formation of mutagenic 4-hydroxy-estradiol, (2) by inhibiting the bioactivation of procarcinogens, and (3) by reducing drug-resistance. Surprisingly, few steroids were reported as inhibitors of CYP1B1. From a screening performed with 90 steroid derivatives, we identified thioestrone (B19) as an inhibitor (IC50=3.4μM) of CYP1B1. Molecular modeling studies showed that the 3-SH group of B19 is closer (3.36Å) to the iron atom of the heme system than the 3-OH group of enzyme substrates estrone and estradiol (4.26Å and 3.58Å, respectively). B19 also produced a better docking GOLD score that correlated with the inhibitory results obtained. The estrane derivative B19 represents an interesting lead compound that can be easily modified to extend the structure-activity relationship study and to provide a next generation of more powerful CYP1B1 inhibitors.
Collapse
|
17
|
Bodnár B, Mernyák E, Wölfling J, Schneider G, Herman BE, Szécsi M, Sinka I, Zupkó I, Kupihár Z, Kovács L. Synthesis and Biological Evaluation of Triazolyl 13α-Estrone-Nucleoside Bioconjugates. Molecules 2016; 21:molecules21091212. [PMID: 27626395 PMCID: PMC6273310 DOI: 10.3390/molecules21091212] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/02/2016] [Accepted: 09/06/2016] [Indexed: 02/03/2023] Open
Abstract
2′-Deoxynucleoside conjugates of 13α-estrone were synthesized by applying the copper-catalyzed alkyne–azide click reaction (CuAAC). For the introduction of the azido group the 5′-position of the nucleosides and a propargyl ether functional group on the 3-hydroxy group of 13α-estrone were chosen. The best yields were realized in our hands when the 3′-hydroxy groups of the nucleosides were protected by acetyl groups and the 5′-hydroxy groups were modified by the tosyl–azide exchange method. The commonly used conditions for click reaction between the protected-5′-azidonucleosides and the steroid alkyne was slightly modified by using 1.5 equivalent of Cu(I) catalyst. All the prepared conjugates were evaluated in vitro by means of MTT assays for antiproliferative activity against a panel of human adherent cell lines (HeLa, MCF-7 and A2780) and the potential inhibitory activity of the new conjugates on human 17β-hydroxysteroid dehydrogenase 1 (17β-HSD1) was investigated via in vitro radiosubstrate incubation. Some protected conjugates displayed moderate antiproliferative properties against a panel of human adherent cancer cell lines (the protected cytidine conjugate proved to be the most potent with IC50 value of 9 μM). The thymidine conjugate displayed considerable 17β-HSD1 inhibitory activity (IC50 = 19 μM).
Collapse
Affiliation(s)
- Brigitta Bodnár
- Department of Medicinal Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary.
| | - Erzsébet Mernyák
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary.
| | - János Wölfling
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary.
| | - Gyula Schneider
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary.
| | - Bianka Edina Herman
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, H-6720 Szeged, Hungary.
| | - Mihály Szécsi
- 1st Department of Medicine, University of Szeged, Korányi fasor 8-10, H-6720 Szeged, Hungary.
| | - Izabella Sinka
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary.
| | - István Zupkó
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary.
| | - Zoltán Kupihár
- Department of Medicinal Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary.
| | - Lajos Kovács
- Department of Medicinal Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary.
| |
Collapse
|
18
|
Szabó J, Pataki Z, Wölfling J, Schneider G, Bózsity N, Minorics R, Zupkó I, Mernyák E. Synthesis and biological evaluation of 13α-estrone derivatives as potential antiproliferative agents. Steroids 2016; 113:14-21. [PMID: 27263437 DOI: 10.1016/j.steroids.2016.05.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 05/25/2016] [Accepted: 05/31/2016] [Indexed: 11/28/2022]
Abstract
13α-Estrone derivatives containing various substituents on C-3 and C-17 were synthesized, and evaluated by means of MTT assays for in vitro antiproliferative activity against a panel of human adherent cancer cell lines (HeLa, MCF-7, A2780 and A431). Compounds with N-benzyltriazolylmethoxy moieties on C-3 proved to be more potent than their 3-hydroxy or 3-ether counterparts. Some triazoles exerted substantial cytostatic effects against particular tumor cell lines, with submicromolar IC50 values.
Collapse
Affiliation(s)
- Johanna Szabó
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Zoltán Pataki
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - János Wölfling
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Gyula Schneider
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Noémi Bózsity
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Renáta Minorics
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - István Zupkó
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary.
| | - Erzsébet Mernyák
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary.
| |
Collapse
|
19
|
Higuchi T, Endo M, Hanamura T, Gohno T, Niwa T, Yamaguchi Y, Horiguchi J, Hayashi SI. Contribution of Estrone Sulfate to Cell Proliferation in Aromatase Inhibitor (AI) -Resistant, Hormone Receptor-Positive Breast Cancer. PLoS One 2016; 11:e0155844. [PMID: 27228187 PMCID: PMC4882040 DOI: 10.1371/journal.pone.0155844] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 05/05/2016] [Indexed: 12/22/2022] Open
Abstract
Aromatase inhibitors (AIs) effectively treat hormone receptor-positive postmenopausal breast cancer, but some patients do not respond to treatment or experience recurrence. Mechanisms of AI resistance include ligand-independent activation of the estrogen receptor (ER) and signaling via other growth factor receptors; however, these do not account for all forms of resistance. Here we present an alternative mechanism of AI resistance. We ectopically expressed aromatase in MCF-7 cells expressing green fluorescent protein as an index of ER activity. Aromatase-overexpressing MCF-7 cells were cultured in estrogen-depleted medium supplemented with testosterone and the AI, letrozole, to establish letrozole-resistant (LR) cell lines. Compared with parental cells, LR cells had higher mRNA levels of steroid sulfatase (STS), which converts estrone sulfate (E1S) to estrone, and the organic anion transporter peptides (OATPs), which mediate the uptake of E1S into cells. LR cells proliferated more in E1S-supplemented medium than did parental cells, and LR proliferation was effectively inhibited by an STS inhibitor in combination with letrozole and by ER-targeting drugs. Analysis of ER-positive primary breast cancer tissues showed a significant correlation between the increases in the mRNA levels of STS and the OATPs in the LR cell lines, which supports the validity of this AI-resistant model. This is the first study to demonstrate the contribution of STS and OATPs in E1S metabolism to the proliferation of AI-resistant breast cancer cells. We suggest that E1S metabolism represents a new target in AI-resistant breast cancer treatment.
Collapse
Affiliation(s)
- Toru Higuchi
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Department of Visceral and Thoracic Organ Surgery, Graduated School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Megumi Endo
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
| | - Toru Hanamura
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Tatsuyuki Gohno
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
| | - Toshifumi Niwa
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
| | - Yuri Yamaguchi
- Research Institute for Clinical Oncology, Saitama Cancer Center, Ina, Saitama, Japan
| | - Jun Horiguchi
- Department of Visceral and Thoracic Organ Surgery, Graduated School of Medicine, Gunma University, Maebashi, Gunma, Japan
| | - Shin-ichi Hayashi
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Center for Regulatory Epi genome and Diseases, Graduate School of Medicine, Tohoku University, Sendai, Niyagi, Japan
| |
Collapse
|
20
|
Current knowledge of the multifunctional 17β-hydroxysteroid dehydrogenase type 1 (HSD17B1). Gene 2016; 588:54-61. [PMID: 27102893 DOI: 10.1016/j.gene.2016.04.031] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 02/10/2016] [Accepted: 04/11/2016] [Indexed: 02/06/2023]
Abstract
At the late 1940s, 17β-HSD1 was discovered as the first member of the 17β-HSD family with its gene cloned. The three-dimensional structure of human 17β-HSD1 is the first example of any human steroid converting enzyme. The human enzyme's structure and biological function have thus been studied extensively in the last two decades. In humans, the enzyme is expressed in placenta, ovary, endometrium and breast. The high activity of estrogen activation provides the basis of 17β-HSD1's implication in estrogen-dependent diseases, such as breast cancer, endometriosis and non-small cell lung carcinomas. Its dual function in estrogen activation and androgen inactivation has been revealed in molecular and breast cancer cell levels, significantly stimulating the proliferation of such cells. The enzyme's overexpression in breast cancer was demonstrated by clinical samples. Inhibition of human 17β-HSD1 led to xenograft tumor shrinkage. Unfortunately, through decades of studies, there is still no drug using the enzyme's inhibitors available. This is due to the difficulty to get rid of the estrogenic activity of its inhibitors, which are mostly estrogen analogues. New non-steroid inhibitors for the enzyme provide new hope for non-estrogenic inhibitors of the enzyme.
Collapse
|
21
|
Szabó J, Bacsa I, Wölfling J, Schneider G, Zupkó I, Varga M, Herman BE, Kalmár L, Szécsi M, Mernyák E. Synthesis and in vitro pharmacological evaluation of N-[(1-benzyl-1,2,3-triazol-4-yl)methyl]-carboxamides on d-secoestrone scaffolds. J Enzyme Inhib Med Chem 2015; 31:574-9. [PMID: 26360618 DOI: 10.3109/14756366.2015.1050008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
An efficient synthesis of several N-[(1-benzyl-1,2,3-triazol-4-yl)methyl]carboxamides in the 13β- and 13α-d-secoestrone series is reported. Novel triazoles were synthesized via the Cu(I)-catalyzed azide-alkyne cycloaddition of steroidal alkynyl carboxamides and p-substituted benzyl azides. Each of the products was evaluated in vitro by means of MTT assays for antiproliferative activity against a panel of human adherent cancer cell lines (HeLa, MCF-7, A431 and A2780). Some of them exhibited activities similar to those of the reference agent cisplatin. On change of the substitution pattern of the benzyl group of the azide, great differences in the cell growth-inhibitory properties were observed. The p-alkylbenzyl-substituted triazoles selectively exerted high cytostatic action against A2780 cells, with IC50 values of 1 µM. We investigated the potential inhibitory action exerted on the human 17β-HSD1 activity of the new secosteroids. Three triazoles effectively suppressed the estrone to 17β-estradiol conversion with IC50 values in low micromolar range.
Collapse
Affiliation(s)
- Johanna Szabó
- a Department of Organic Chemistry , University of Szeged , Szeged , Hungary
| | - Ildikó Bacsa
- a Department of Organic Chemistry , University of Szeged , Szeged , Hungary
| | - János Wölfling
- a Department of Organic Chemistry , University of Szeged , Szeged , Hungary
| | - Gyula Schneider
- a Department of Organic Chemistry , University of Szeged , Szeged , Hungary
| | - István Zupkó
- b Department of Pharmacodynamics and Biopharmacy , University of Szeged , Szeged , Hungary
| | - Mónika Varga
- c Cereal Research Non-Profit Ltd. , Szeged , Hungary
| | - Bianka E Herman
- d 1st Department of Medicine , University of Szeged, Szeged , Hungary , and
| | - László Kalmár
- e Department of Obstetrics and Gynecology , University of Szeged , Szeged , Hungary
| | - Mihály Szécsi
- d 1st Department of Medicine , University of Szeged, Szeged , Hungary , and
| | - Erzsébet Mernyák
- a Department of Organic Chemistry , University of Szeged , Szeged , Hungary
| |
Collapse
|
22
|
Srungboonmee K, Songtawee N, Monnor T, Prachayasittikul V, Nantasenamat C. Probing the origins of 17β-hydroxysteroid dehydrogenase type 1 inhibitory activity via QSAR and molecular docking. Eur J Med Chem 2015; 96:231-7. [DOI: 10.1016/j.ejmech.2015.04.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 04/07/2015] [Accepted: 04/09/2015] [Indexed: 11/29/2022]
|
23
|
Mori T, Ito F, Matsushima H, Takaoka O, Koshiba A, Tanaka Y, Kusuki I, Kitawaki J. Dienogest reduces HSD17β1 expression and activity in endometriosis. J Endocrinol 2015; 225:69-76. [PMID: 25767055 DOI: 10.1530/joe-15-0052] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2015] [Indexed: 11/08/2022]
Abstract
Endometriosis is an estrogen-dependent disease. Abnormally biosynthesized estrogens in endometriotic tissues induce the growth of the lesion and worsen endometriosis-associated pelvic pain. Dienogest (DNG), a selective progesterone receptor agonist, is widely used to treat endometriosis and efficiently relieves the symptoms. However, its pharmacological action remains unknown. In this study, we elucidated the effect of DNG on enzymes involved in local estrogen metabolism in endometriosis. Surgically obtained specimens of 23 ovarian endometriomas (OE) and their homologous endometrium (EE), ten OE treated with DNG (OE w/D), and 19 normal endometria without endometriosis (NE) were analyzed. Spheroid cultures of stromal cells (SCs) were treated with DNG and progesterone. The expression of aromatase, 17β-hydroxysteroid dehydrogenase 1 (HSD17β1), HSD17β2, HSD17β7, HSD17β12, steroid sulfatase (STS), and estrogen sulfotransferase (EST) was evaluated by real-time quantitative PCR. The activity and protein level of HSD17β1 were measured with an enzyme assay using radiolabeled estrogens and immunohistochemistry respectively. OESCs showed increased expression of aromatase, HSD17β1, STS, and EST, along with decreased HSD17β2 expression, when compared with stromal cells from normal endometria without endometriosis (NESCs) (P<0.01) or stromal cells from homologous endometrium (EESCs) (P<0.01). In OESCs, DNG inhibited HSD17β1 expression and enzyme activity at 10(-7) M (P<0.01). Results of immunohistochemical analysis displayed reduced HSD17β1 staining intensity in OE w/D (P<0.05). In conclusion, DNG exerts comprehensive inhibition of abnormal estrogen production through inhibition of aromatase and HSD17β1, contributing to a therapeutic effect of DNG on endometriosis.
Collapse
Affiliation(s)
- Taisuke Mori
- Department of Obstetrics and Gynecology Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Fumitake Ito
- Department of Obstetrics and Gynecology Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Hiroshi Matsushima
- Department of Obstetrics and Gynecology Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Osamu Takaoka
- Department of Obstetrics and Gynecology Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Akemi Koshiba
- Department of Obstetrics and Gynecology Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yukiko Tanaka
- Department of Obstetrics and Gynecology Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Izumi Kusuki
- Department of Obstetrics and Gynecology Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Jo Kitawaki
- Department of Obstetrics and Gynecology Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
24
|
Identification of fused 16β,17β-oxazinone-estradiol derivatives as a new family of non-estrogenic 17β-hydroxysteroid dehydrogenase type 1 inhibitors. Eur J Med Chem 2015; 93:470-80. [DOI: 10.1016/j.ejmech.2015.01.059] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/28/2015] [Accepted: 01/30/2015] [Indexed: 01/19/2023]
|
25
|
Maltais R, Ayan D, Trottier A, Barbeau X, Lagüe P, Bouchard JE, Poirier D. Discovery of a Non-Estrogenic Irreversible Inhibitor of 17β-Hydroxysteroid Dehydrogenase Type 1 from 3-Substituted-16β-(m-carbamoylbenzyl)-estradiol Derivatives. J Med Chem 2013; 57:204-22. [DOI: 10.1021/jm401639v] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- René Maltais
- Laboratory
of Medicinal Chemistry, Oncology and Nephrology Unit, CHU de Québec—Research
Center (CHUL, T4-42) and Faculty of Medicine, Laval University, Québec
City, Québec G1V
4G2, Canada
| | - Diana Ayan
- Laboratory
of Medicinal Chemistry, Oncology and Nephrology Unit, CHU de Québec—Research
Center (CHUL, T4-42) and Faculty of Medicine, Laval University, Québec
City, Québec G1V
4G2, Canada
| | - Alexandre Trottier
- Laboratory
of Medicinal Chemistry, Oncology and Nephrology Unit, CHU de Québec—Research
Center (CHUL, T4-42) and Faculty of Medicine, Laval University, Québec
City, Québec G1V
4G2, Canada
| | - Xavier Barbeau
- Département
de Chimie, Institut de Biologie Intégrative et Des Systèmes
(IBIS), and Centre de Recherche sur la Fonction, la Structure et l’Ingénierie
des Protéines (PROTEO), Université Laval, Québec City, Québec G1V 4G2, Canada
| | - Patrick Lagüe
- Département
de Biochimie Microbiologie et Bio-informatique, Institut de Biologie
Intégrative et des Systèmes (IBIS), and Centre de Recherche
sur la Fonction, la Structure et l’Ingénierie des Protéines
(PROTEO), Université Laval, Québec City, Québec G1V 4G2, Canada
| | - Jean-Emmanuel Bouchard
- Laboratory
of Medicinal Chemistry, Oncology and Nephrology Unit, CHU de Québec—Research
Center (CHUL, T4-42) and Faculty of Medicine, Laval University, Québec
City, Québec G1V
4G2, Canada
| | - Donald Poirier
- Laboratory
of Medicinal Chemistry, Oncology and Nephrology Unit, CHU de Québec—Research
Center (CHUL, T4-42) and Faculty of Medicine, Laval University, Québec
City, Québec G1V
4G2, Canada
| |
Collapse
|
26
|
Cysteine-10 on 17 β -Hydroxysteroid Dehydrogenase 1 Has Stabilizing Interactions in the Cofactor Binding Region and Renders Sensitivity to Sulfhydryl Modifying Chemicals. Int J Cell Biol 2013; 2013:769536. [PMID: 24348564 PMCID: PMC3855964 DOI: 10.1155/2013/769536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 09/15/2013] [Indexed: 11/29/2022] Open
Abstract
17β-Hydroxysteroid dehydrogenase type 1 (17β-HSD1) catalyzes the conversion of estrone to the potent estrogen estradiol. 17β-HSD1 is highly expressed in breast and ovary tissues and represents a prognostic marker for the tumor progression and survival of patients with breast cancer and other estrogen-dependent tumors. Therefore, the enzyme is considered a promising drug target against estrogen-dependent cancers. For the development of novel inhibitors, an improved understanding of the structure-function relationships is essential. In the present study, we examined the role of a cysteine residue, Cys10, in the Rossmann-fold NADPH binding region, for 17β-HSD1 function and tested the sensitivity towards sulfhydryl modifying chemicals. 3D structure modeling revealed important interactions of Cys10 with residues involved in the stabilization of amino acids of the NADPH binding pocket. Analysis of enzyme activity revealed that 17β-HSD1 was irreversibly inhibited by the sulfhydryl modifying agents N-ethylmaleimide (NEM) and dithiocarbamates. Preincubation with increasing concentrations of NADPH protected 17β-HSD1 from inhibition by these chemicals. Cys10Ser mutant 17β-HSD1 was partially protected from inhibition by NEM and dithiocarbamates, emphasizing the importance of Cys10 in the cofactor binding region. Substitution of Cys10 with serine resulted in a decreased protein half-life, without significantly altering kinetic properties. Despite the fact that Cys10 on 17β-HSD1 seems to have limited potential as a target for new enzyme inhibitors, the present study provides new insight into the structure-function relationships of this enzyme.
Collapse
|
27
|
Secky L, Svoboda M, Klameth L, Bajna E, Hamilton G, Zeillinger R, Jäger W, Thalhammer T. The sulfatase pathway for estrogen formation: targets for the treatment and diagnosis of hormone-associated tumors. JOURNAL OF DRUG DELIVERY 2013; 2013:957605. [PMID: 23476785 PMCID: PMC3586502 DOI: 10.1155/2013/957605] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 12/17/2012] [Indexed: 12/15/2022]
Abstract
The extragonadal synthesis of biological active steroid hormones from their inactive precursors in target tissues is named "intracrinology." Of particular importance for the progression of estrogen-dependent cancers is the in situ formation of the biological most active estrogen, 17beta-estradiol (E2). In cancer cells, conversion of inactive steroid hormone precursors to E2 is accomplished from inactive, sulfated estrogens in the "sulfatase pathway" and from androgens in the "aromatase pathway." Here, we provide an overview about expression and function of enzymes of the "sulfatase pathway," particularly steroid sulfatase (STS) that activates estrogens and estrogen sulfotransferase (SULT1E1) that converts active estrone (E1) and other estrogens to their inactive sulfates. High expression of STS and low expression of SULT1E1 will increase levels of active estrogens in malignant tumor cells leading to the stimulation of cell proliferation and cancer progression. Therefore, blocking the "sulfatase pathway" by STS inhibitors may offer an attractive strategy to reduce levels of active estrogens. STS inhibitors either applied in combination with aromatase inhibitors or as novel, dual aromatase-steroid sulfatase inhibiting drugs are currently under investigation. Furthermore, STS inhibitors are also suitable as enzyme-based cancer imaging agents applied in the biomedical imaging technique positron emission tomography (PET) for cancer diagnosis.
Collapse
Affiliation(s)
- Lena Secky
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Martin Svoboda
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Lukas Klameth
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Ludwig Boltzmann Cluster Translational Oncology, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Erika Bajna
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Gerhard Hamilton
- Ludwig Boltzmann Cluster Translational Oncology, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Robert Zeillinger
- Ludwig Boltzmann Cluster Translational Oncology, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Walter Jäger
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, 1090 Vienna, Austria
| | - Theresia Thalhammer
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
28
|
Ayan D, Maltais R, Roy J, Poirier D. A new nonestrogenic steroidal inhibitor of 17β-hydroxysteroid dehydrogenase type I blocks the estrogen-dependent breast cancer tumor growth induced by estrone. Mol Cancer Ther 2012; 11:2096-104. [PMID: 22914440 DOI: 10.1158/1535-7163.mct-12-0299] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
17β-Hydroxysteroid dehydrogenase type 1 (17β-HSD1) converts estrone (E1) into estradiol (E2) and is expressed in many steroidogenic tissues and breast cancer cell lines. Because the potent estrogen E2 stimulates the growth and development of hormone-dependent diseases, inhibition of the final step of E2 synthesis is considered a promising strategy for the treatment of breast cancer. On the basis of our previous study identifying 16β-(m-carbamoylbenzyl)-E2 (CC-156) as a lead compound for the inhibition of 17β-HSD1, we conducted a number of structural modifications to reduce its undesired residual estrogenic activity. The steroid derivative PBRM [3-(2-bromoethyl)-16β-(m-carbamoylbenzyl)-17β-hydroxy-1,3,5(10)-estratriene] emerged as a potent inhibitor of 17β-HSD1 with an IC(50) value of 68 nmol/L for the transformation of E1 into E2. When tested in the estrogen-sensitive breast cancer cell line T-47D and in mice, PBRM showed no estrogenic activity in the range of concentrations tested. Furthermore, with the purpose of evaluating the bioavailability of PBRM and CC-156 injected subcutaneously (2.3 mg/kg), we measured their plasmatic concentrations as a function of time, calculated the area under the curve (AUC(0-12h)) and showed a significant improvement for PBRM (772 ng*h/mL) compared with CC-156 (445 ng*h/mL). We next tested the in vivo efficiency of PBRM on the T-47D xenograft tumor model in female ovariectomized athymic nude mice. After a treatment with PBRM, tumor sizes in mice stimulated with exogenous E1 were completely reduced at the control group level (without E1 treatment). As a conclusion, PBRM is a promising nonestrogenic inhibitor of 17β-HSD1 for the treatment of estrogen-dependent diseases such as breast cancer.
Collapse
Affiliation(s)
- Diana Ayan
- Laboratory of Medicinal Chemistry, CHUQ (CHUL)-Research Center, 2705 Laurier Boulevard, Quebec G1V 4G2, Canada
| | | | | | | |
Collapse
|
29
|
Ayan D, Maltais R, Poirier D. Identification of a 17β-hydroxysteroid dehydrogenase type 10 steroidal inhibitor: a tool to investigate the role of type 10 in Alzheimer's disease and prostate cancer. ChemMedChem 2012; 7:1181-4. [PMID: 22674910 DOI: 10.1002/cmdc.201200129] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Indexed: 11/09/2022]
Affiliation(s)
- Diana Ayan
- Laboratory of Medicinal Chemistry, Endocrinology and Genomic Unit, CHUQ-CHUL Research Center, 2705 Boulevard Laurier, Québec G1V 4G2, Canada
| | | | | |
Collapse
|
30
|
Rotinen M, Villar J, Encío I. Regulation of 17β-hydroxysteroid dehydrogenases in cancer: regulating steroid receptor at pre-receptor stage. J Physiol Biochem 2012; 68:461-73. [DOI: 10.1007/s13105-012-0155-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 02/07/2012] [Indexed: 11/27/2022]
|
31
|
Yoshimoto FK, Desilets MC, Auchus RJ. Synthesis of halogenated pregnanes, mechanistic probes of steroid hydroxylases CYP17A1 and CYP21A2. J Steroid Biochem Mol Biol 2012; 128:38-50. [PMID: 22001566 PMCID: PMC3306177 DOI: 10.1016/j.jsbmb.2011.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 09/22/2011] [Accepted: 09/24/2011] [Indexed: 01/11/2023]
Abstract
The human steroidogenic cytochromes P450 CYP17A1 (P450c17, 17α-hydroxylase/17,20-lyase) and CYP21A2 (P450c21, 21-hydroxylase) are required for the biosynthesis of androgens, glucocorticoids, and mineralocorticoids. Both enzymes hydroxylate progesterone at adjacent, distal carbon atoms and show limited tolerance for substrate modification. Halogenated substrate analogs have been employed for many years to probe cytochrome P450 catalysis and to block sites of reactivity, particularly for potential drugs. Consequently, we developed efficient synthetic approaches to introducing one or more halogen atom to the 17- and 21-positions of progesterone and pregnenolone. In particular, novel 21,21,21-tribromoprogesterone and 21,21,21-trichloroprogesterone were synthesized using the nucleophilic addition of either bromoform or chloroform anion onto an aldehyde precursor as the key step to introduce the trihalomethyl moieties. When incubated with microsomes from yeast expressing human CYP21A2 or CYP17A1 with P450-oxidoreductase, CYP21A2 metabolized 17-fluoroprogesterone to a single product, whereas incubations with CYP17A1 gave no products. Halogenated steroids provide a robust system for exploring the substrate tolerance and catalytic plasticity of human steroid hydroxylases.
Collapse
Affiliation(s)
- Francis K Yoshimoto
- Division of Endocrinology & Metabolism, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390-8857, United States
| | | | | |
Collapse
|
32
|
Ayan D, Roy J, Maltais R, Poirier D. Impact of estradiol structural modifications (18-methyl and/or 17-hydroxy inversion of configuration) on the in vitro and in vivo estrogenic activity. J Steroid Biochem Mol Biol 2011; 127:324-30. [PMID: 21827856 DOI: 10.1016/j.jsbmb.2011.07.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 07/15/2011] [Accepted: 07/26/2011] [Indexed: 12/14/2022]
Abstract
It is well recognized that the majority of breast cancers are initially hormone-dependent and that 17β-estradiol (17β-E2) plays a crucial role in their development and progression. For this reason, using a compound able to block a specific enzyme involved in the last steps of the biosynthesis of 17β-E2 remains a rational way to treat estrogen-dependent diseases such as breast cancer. The present study describes the biological in vitro and in vivo evaluation of a structural modification (inversion of C18-methyl group at position 13 from β to α face) of 17β-E2 (1) and 17α-estradiol (17α-E2; 2). The two epimers 18-epi-17β-E2 (3) and 18-epi-17α-E2 (4) were obtained in two chemical steps by inversion of the C18-methyl of estrone using 1,2-phenylendiamine in refluxing acetic acid and reduction of ketone at position C17 with LiAlH(4). The new E2 isomers were tested on estrogen-sensitive cell lines (MCF-7 and T-47D), on estrogen-sensitive tissues (uterus and vagina of mice) and on estrogen receptor (ER) to determine their estrogenic potency relatively to natural estrogen 17β-E2 (1). The results show that 18-epi-17β-E2 (3) possesses the lower affinity for ER (RBA = 1.2%), the lower estrogenicity on estrogen-sensitive cells (1000 folds less estrogenic than 17β-E2 in MCF-7) and no uterotrophic (estrogenic) activity when tested on mice. In fact, we observed the following order of estrogenicity: 18-epi-17β-E2 (3)<18-epi-17α-E2 (4) << 17α-E2 (2)17β-E2 (1). These results suggest that the inversion of C18-methyl of natural 17β-E2 scaffold could be a useful strategy to decrease the estrogenicity of E2 derivatives used as enzyme inhibitors in the context of a treatment of estrogen-dependent diseases.
Collapse
Affiliation(s)
- Diana Ayan
- Laboratory of Medicinal Chemistry, CHUQ, CHUL-Research Center, Endocrinology and Genomic Unit, and Laval University, Faculty of Medicine, Quebec, Quebec G1V 4G2, Canada
| | | | | | | |
Collapse
|
33
|
Maltais R, Ayan D, Poirier D. Crucial Role of 3-Bromoethyl in Removing the Estrogenic Activity of 17β-HSD1 Inhibitor 16β-(m-Carbamoylbenzyl)estradiol. ACS Med Chem Lett 2011; 2:678-681. [PMID: 21927646 PMCID: PMC3174009 DOI: 10.1021/ml200093v] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 07/17/2011] [Indexed: 01/11/2023] Open
Abstract
![]()
17β-Hydroxysteroid dehydrogenase type 1 (17β-HSD1) represents a promising therapeutic target for breast cancer treatment. To reduce the undesirable estrogenic activity of potent 17β-HSD1 inhibitor 16β-(m-carbamoylbenzyl)estradiol (1) (IC50 = 27 nM), a series of analogues with a small functionalized side chain at position 3 were synthesized and tested. The 3-(2-bromoethyl)-16β-(m-carbamoylbenzyl)-estra-1,3,5(10)-trien-17β-ol (5) was found to be a potent inhibitor (IC50 = 68 nM) for the transformation of estrone (E1) into estradiol (E2) and, most importantly, did not stimulate the proliferation of estrogen-sensitive MCF-7 cells, suggesting no estrogenic activity. From these results, the crucial role of a bromoalkyl side chain at carbon 3 was identified for the first time. Thus, this new inhibitor represents a good candidate with an interesting profile suitable for further studies including pharmacokinetic and in vivo studies.
Collapse
Affiliation(s)
- René Maltais
- Laboratory of Medicinal Chemistry, Endocrinology and Genomic Unit, CHUQ (CHUL) − Research Center and Laval University, Quebec (Quebec) G1V 4G2, Canada
| | - Diana Ayan
- Laboratory of Medicinal Chemistry, Endocrinology and Genomic Unit, CHUQ (CHUL) − Research Center and Laval University, Quebec (Quebec) G1V 4G2, Canada
| | - Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Genomic Unit, CHUQ (CHUL) − Research Center and Laval University, Quebec (Quebec) G1V 4G2, Canada
| |
Collapse
|
34
|
Ouellet É, Ayan D, Poirier D. Synthesis and preliminary evaluation of a modified estradiol-core bearing a fused γ-lactone as non-estrogenic inhibitor of 17β-hydroxysteroid dehydrogenase type 1. Bioorg Med Chem Lett 2011; 21:5510-3. [DOI: 10.1016/j.bmcl.2011.06.110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 06/24/2011] [Indexed: 10/18/2022]
|
35
|
Byrns MC, Jin Y, Penning TM. Inhibitors of type 5 17β-hydroxysteroid dehydrogenase (AKR1C3): overview and structural insights. J Steroid Biochem Mol Biol 2011; 125:95-104. [PMID: 21087665 PMCID: PMC3047600 DOI: 10.1016/j.jsbmb.2010.11.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 10/27/2010] [Accepted: 11/05/2010] [Indexed: 12/27/2022]
Abstract
There is considerable interest in the development of an inhibitor of aldo-keto reductase (AKR) 1C3 (type 5 17β-hydroxysteroid dehydrogenase and prostaglandin F synthase) as a potential therapeutic for both hormone-dependent and hormone-independent cancers. AKR1C3 catalyzes the reduction of 4-androstene-3,17-dione to testosterone and estrone to 17β-estradiol in target tissues, which will promote the proliferation of hormone dependent prostate and breast cancers, respectively. AKR1C3 also catalyzes the reduction of prostaglandin (PG) H(2) to PGF(2α) and PGD(2) to 9α,11β-PGF(2), which will limit the formation of anti-proliferative prostaglandins, including 15-deoxy-Δ(12,14)-PGJ(2), and contribute to proliferative signaling. AKR1C3 is overexpressed in a wide variety of cancers, including breast and prostate cancer. An inhibitor of AKR1C3 should not inhibit the closely related isoforms AKR1C1 and AKR1C2, as they are involved in other key steroid hormone biotransformations in target tissues. Several structural leads have been explored as inhibitors of AKR1C3, including non-steroidal anti-inflammatory drugs, steroid hormone analogues, flavonoids, cyclopentanes, and benzodiazepines. Inspection of the available crystal structures of AKR1C3 with multiple ligands bound, along with the crystal structures of the other AKR1C isoforms, provides a structural basis for the rational design of isoform specific inhibitors of AKR1C3. We find that there are subpockets involved in ligand binding that are considerably different in AKR1C3 relative to the closely related AKR1C1 or AKR1C2 isoforms. These pockets can be used to further improve the binding affinity and selectivity of the currently available AKR1C3 inhibitors. Article from the special issue on Targeted Inhibitors.
Collapse
Affiliation(s)
| | | | - Trevor M. Penning
- Corresponding author. Tel.: +1 215 898 9445; fax: +1 215 573 2236. (T.M. Penning)
| |
Collapse
|
36
|
Penning TM. Human hydroxysteroid dehydrogenases and pre-receptor regulation: insights into inhibitor design and evaluation. J Steroid Biochem Mol Biol 2011; 125:46-56. [PMID: 21272640 PMCID: PMC3104102 DOI: 10.1016/j.jsbmb.2011.01.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 01/18/2011] [Accepted: 01/18/2011] [Indexed: 11/16/2022]
Abstract
Hydroxysteroid dehydrogenases (HSDs) represent a major class of NAD(P)(H) dependent steroid hormone oxidoreductases involved in the pre-receptor regulation of hormone action. This is achieved by HSDs working in pairs so that they can interconvert ketosteroids with hydroxysteroids resulting in a change in ligand potency for nuclear receptors. HSDs belong to two protein superfamilies the aldo-keto reductases and the short-chain dehydrogenase/reductases. In humans, many of the important enzymes have been thoroughly characterized including the elucidation of their three-dimensional structures. Because these enzymes play fundamental roles in steroid hormone action they can be considered to be drug targets for a variety of steroid driven diseases, e.g. metabolic syndrome and obesity, inflammation, and hormone dependent malignancies of the endometrium, prostate and breast. This article will review how fundamental knowledge of these enzymes can be exploited in the development of isoform specific HSD inhibitors from both protein superfamilies. Article from the Special issue on Targeted Inhibitors.
Collapse
Affiliation(s)
- Trevor M Penning
- Center of Excellence in Environmental Toxicology, Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6084, USA.
| |
Collapse
|