1
|
Templ J, Schnürch M. Allylation of C-, N-, and O-Nucleophiles via a Mechanochemically-Driven Tsuji-Trost Reaction Suitable for Late-Stage Modification of Bioactive Molecules. Angew Chem Int Ed Engl 2024; 63:e202314637. [PMID: 37931225 PMCID: PMC10952285 DOI: 10.1002/anie.202314637] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/08/2023]
Abstract
We present the first solvent-free, mechanochemical protocol for a palladium-catalyzed Tsuji-Trost allylation. This approach features exceptionally low catalyst loadings (0.5 mol %), short reaction times (<90 min), and a simple setup, eliminating the need for air or moisture precautions, making the process highly efficient and environmentally benign. We introduce solid, nontoxic, and easy-to-handle allyl trimethylammonium salts as valuable alternative to volatile or hazardous reagents. Our approach enables the allylation of various O-, N-, and C-nucleophiles in yields up to 99 % even for structurally complex bioactive compounds, owing to its mild conditions and exceptional functional group tolerance.
Collapse
Affiliation(s)
- Johanna Templ
- Institute of Applied Synthetic Chemistry, TU WienGetreidemarkt 9/E1631060ViennaAustria
| | - Michael Schnürch
- Institute of Applied Synthetic Chemistry, TU WienGetreidemarkt 9/E1631060ViennaAustria
| |
Collapse
|
2
|
Templ J, Schnürch M. Allylation of C-, N-, and O-Nucleophiles via a Mechanochemically-Driven Tsuji-Trost Reaction Suitable for Late-Stage Modification of Bioactive Molecules. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 136:e202314637. [PMID: 38516646 PMCID: PMC10953357 DOI: 10.1002/ange.202314637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Indexed: 03/23/2024]
Abstract
We present the first solvent-free, mechanochemical protocol for a palladium-catalyzed Tsuji-Trost allylation. This approach features exceptionally low catalyst loadings (0.5 mol %), short reaction times (<90 min), and a simple setup, eliminating the need for air or moisture precautions, making the process highly efficient and environmentally benign. We introduce solid, nontoxic, and easy-to-handle allyl trimethylammonium salts as valuable alternative to volatile or hazardous reagents. Our approach enables the allylation of various O-, N-, and C-nucleophiles in yields up to 99 % even for structurally complex bioactive compounds, owing to its mild conditions and exceptional functional group tolerance.
Collapse
Affiliation(s)
- Johanna Templ
- Institute of Applied Synthetic Chemistry, TU WienGetreidemarkt 9/E1631060ViennaAustria
| | - Michael Schnürch
- Institute of Applied Synthetic Chemistry, TU WienGetreidemarkt 9/E1631060ViennaAustria
| |
Collapse
|
3
|
Dohle W, Asiki H, Gruchot W, Foster PA, Sahota HK, Bai R, Christensen KE, Hamel E, Potter BVL. 2-Difluoromethoxy-Substituted Estratriene Sulfamates: Synthesis, Antiproliferative SAR, Antitubulin Activity, and Steroid Sulfatase Inhibition. ChemMedChem 2022; 17:e202200408. [PMID: 36109340 PMCID: PMC9742152 DOI: 10.1002/cmdc.202200408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/14/2022] [Indexed: 01/14/2023]
Abstract
2-Difluoromethoxyestratriene derivatives were designed to improve potency and in vivo stability of the drug candidate 2-methoxyestradiol (2ME2). Compound evaluation in vitro against the proliferation of MCF-7 and MDA MB-231 breast cancer cells, as inhibitors of tubulin polymerisation and also steroid sulfatase (STS) both in cell lysates and in whole cells, showed promising activities. In antiproliferative assays 2-difluoromethoxyestradiol was less potent than 2ME2, but its sulfamates were often more potent than their corresponding non-fluorinated analogues. The fluorinated bis-sulfamate is a promising antiproliferative agent in MCF-7 cells (GI50 0.28 μM) vs the known 2-methoxyestradiol-3,17-O,O-bissulfamate (STX140, GI50 0.52 μM), confirming the utility of our approach. Compounds were also evaluated in the NCI 60-cell line panel and the fluorinated bis-sulfamate derivative displayed very good overall activities with a sub-micromolar average GI50 . It was a very potent STS inhibitor in whole JEG-3 cells (IC50 3.7 nM) similar to STX140 (4.2 nM) and additionally interferes with tubulin assembly in vitro and colchicine binding to tubulin. An X-ray study of 2-difluoromethoxy-3-benzyloxyestra-1,3,5(10)-trien-17-one examined conformational aspects of the fluorinated substituent. The known related derivative 2-difluoromethyl-3-sulfamoyloxyestrone was evaluated for STS inhibition in whole JEG-3 cells and showed an excellent IC50 of 55 pM.
Collapse
Affiliation(s)
- Wolfgang Dohle
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Hannah Asiki
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Wojciech Gruchot
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Paul A Foster
- Institute of Metabolism & Systems Research, University of Birmingham, 2nd Floor IBR Tower Edgbaston, Birmingham, B15 2TT, UK
- Centre for Endocrinology, Metabolism and Diabetes, University of Birmingham, Birmingham Health Partners, Birmingham, B15 2TT, UK
| | - Havreen K Sahota
- Institute of Metabolism & Systems Research, University of Birmingham, 2nd Floor IBR Tower Edgbaston, Birmingham, B15 2TT, UK
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD, 21702, USA
| | - Kirsten E Christensen
- Chemical Crystallography, Department of Chemistry, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD, 21702, USA
| | - Barry V L Potter
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| |
Collapse
|
4
|
Kotha S, Solanke BU, Agarwala PK, Kapoor S. Design and synthesis of hetero-steroids via ring-closing metathesis: Biological studies towards in vitro anticancer activity. Steroids 2022; 188:109119. [PMID: 36202314 DOI: 10.1016/j.steroids.2022.109119] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 11/15/2022]
Abstract
Here, we report a synthetic approach to hetero-steroids and also studied their biological activities as anticancer agents. A novel class of oxacycles containing estrone moiety were synthesized in this report. Allyl ether derived from estrone underwent Claisen rearrangement (CR) and again O-allylation and subsequent ring-closure gave A-ring-furan and oxepine fused derivatives in high yields. We used double bond isomerization and ring-closing metathesis (RCM) as key steps to assemble hetero steroids containing a mixture of regio isomers like benzofurans and benzoxepine moieties. The novel benzofuran and benzoxepine-based hybrid steroid derivatives were subjected to in vitro cytotoxicity analysis and were found to exert cancer cell-specific activity.
Collapse
Affiliation(s)
- Sambasivarao Kotha
- Department of Chemistry, Indian Institute of Technology-Bombay, Powai, Mumbai 400076, India.
| | - Balaji U Solanke
- Department of Chemistry, Indian Institute of Technology-Bombay, Powai, Mumbai 400076, India
| | - Prema Kumari Agarwala
- Department of Chemistry, Indian Institute of Technology-Bombay, Powai, Mumbai 400076, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology-Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
5
|
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
|
6
|
C-Ring Oxidized Estrone Acetate Derivatives: Assessment of Antiproliferative Activities and Docking Studies. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12073579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
C-Ring oxidized estrone acetate derivatives as antiproliferative agents were prepared and tested against five cancer cell lines by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Flow cytometry assays to evaluate cell viability and modifications in cell cycle phases and molecular docking research against estrogen receptor α, steroid sulfatase, and 17β-hydroxysteroid dehydrogenase type 1 were performed. 9α-Hydroxy,11β-nitrooxyestrone acetate was the most cytotoxic molecule against hormone-dependent cancer cells. Furthermore, flow cytometry experiments revealed that this 9α-hydroxy,11β-nitrooxy derivative markedly reduced HepaRG cells viability (~92%) after 24 h of treatment. However, 9α-hydroxyestrone acetate led to selective inhibition of HepaRG cells growth, inducing a G0/G1 cycle arrest, and did not originate a proliferation effect on T47-D cancer cells. Docking studies estimated a generally lower affinity of these compounds to estrogen receptor α than predicted for estrone and 17β-estradiol. Therefore, this structural modification can be of interest to develop new anticancer estrane derivatives devoid of estrogenic action.
Collapse
|
7
|
Guin S, Majee D, Samanta S. Unmasking the reverse reactivity of cyclic N-sulfonyl ketimines: multifaceted applications in organic synthesis. Chem Commun (Camb) 2021; 57:9010-9028. [PMID: 34498642 DOI: 10.1039/d1cc03439a] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The chemistry related to the exploration of cyclic N-sulfonyl ketimines and their derivatives has attracted significant attention in the last few decades because of their intriguing structures and properties. They serve broadly as reactive synthons in various reactions to create a diverse set of synthetically and biologically attractive molecules. Furthermore, these moieties, which possess multiple heteroatoms (N, O and S), display or can enhance many biological activities. In the case of synthetic reactions, chemists mainly focus on the chemical manipulation of the highly reactive prochiral CN bond of N-sulfonyl ketimines. Besides their traditional role as electrophiles, N-sulfonyl ketimines possess α-Csp3-H protons, and thus behave as potential carbonucleophiles, where they can undergo several C-X (X = C, N and O) bond-forming reactions with different types of electrophiles under various conditions to form a wide range of fascinating asymmetric and non-asymmetric versions of fused heterocycles, carbocycles, spiro-fused skeletons, pyridines, pyrroles, etc. Herein, we highlight the recent examples from our research work and others covering the scope of cyclic N-sulfonyl ketimines as useful carbonucleophiles. In addition, the detailed mechanistic studies of the above-mentioned reactions are also presented.
Collapse
Affiliation(s)
- Soumitra Guin
- Department of Chemistry, Indian Institute of Technology Indore, Simrol, Indore, 453552, India.
| | - Debashis Majee
- Department of Chemistry, Indian Institute of Technology Indore, Simrol, Indore, 453552, India.
| | - Sampak Samanta
- Department of Chemistry, Indian Institute of Technology Indore, Simrol, Indore, 453552, India.
| |
Collapse
|
8
|
Mernyák E, Bartha S, Kóczán L, Jójárt R, Resch V, Paragi G, Vágvölgyi M, Hunyadi A, Bruszel B, Zupkó I, Minorics R. Microwave-assisted Phospha-Michael addition reactions in the 13α-oestrone series and in vitro antiproliferative properties. J Enzyme Inhib Med Chem 2021; 36:1931-1937. [PMID: 34445919 PMCID: PMC8405091 DOI: 10.1080/14756366.2021.1963241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Microwave-assisted phospha-Michael addition reactions were carried out in the 13α-oestrone series. The exocyclic 16-methylene-17-ketones as α,β-unsaturated ketones were reacted with secondary phosphine oxides as nucleophilic partners. The addition reactions furnished the two tertiary phosphine oxide diastereomers in high yields. The main product was the 16α-isomer. The antiproliferative activities of the newly synthesised organophosphorus compounds against a panel of nine human cancer cell lines were investigated by means of MTT assays. The most potent compound, the diphenylphosphine oxide derivative in the 3-O-methyl-13α-oestrone series (9), exerted selective cell growth-inhibitory activity against UPCI-SCC-131 and T47D cell lines with low micromolar IC50 values. Moreover, it displayed good tumour selectivity property determined against non-cancerous mouse fibroblast cells.
Collapse
Affiliation(s)
- Erzsébet Mernyák
- Department of Organic Chemistry, University of Szeged, Szeged, Hungary
| | - Sándor Bartha
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Szeged, Hungary
| | - Lili Kóczán
- Department of Organic Chemistry, University of Szeged, Szeged, Hungary
| | - Rebeka Jójárt
- Department of Organic Chemistry, University of Szeged, Szeged, Hungary
| | - Vivien Resch
- Department of Medicinal Chemistry, University of Szeged, Szeged, Hungary
| | - Gábor Paragi
- MTA-SZTE Biomimetic Systems Research Group, University of Szeged, Szeged, Hungary.,Institute of Physics, University of Pécs, Pécs, Hungary
| | - Máté Vágvölgyi
- Department of Pharmacognosy, University of Szeged, Szeged, Hungary
| | - Attila Hunyadi
- Department of Pharmacognosy, University of Szeged, Szeged, Hungary
| | - Bella Bruszel
- Department of Medicinal Chemistry, University of Szeged, Szeged, Hungary
| | - István Zupkó
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Szeged, Hungary
| | - Renáta Minorics
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Szeged, Hungary
| |
Collapse
|
9
|
Huang L, Huang J, Nie H, Li Y, Song L, Wu F. Design, synthesis and biological evaluation of combretastatin A-4 sulfamate derivatives as potential anti-cancer agents. RSC Med Chem 2021; 12:1374-1380. [PMID: 34458740 PMCID: PMC8372205 DOI: 10.1039/d0md00372g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 05/17/2021] [Indexed: 01/20/2023] Open
Abstract
A series of combretastatin A-4 (CA-4) sulfamate derivatives were synthesized and their structure-activity relationship on tubulin, arylsulfatase and tumor cell antiproliferation inhibition was studied. Among them, compound 16a showed excellent potency as well as CA-4 under the same conditions against six tumor cells including HTC-116, HeLa, HepG2, MGC803, MKN45 and MCF-7 cells, respectively. Molecular docking revealed that several important hydrogen bond interactions were formed between the sulfamate group of 16a and the colchicine binding site of tubulin and steroid sulfatase respectively. Although compound 16a was less active than CA-4 in regard to its in vitro activity as an inhibitor of tubulin polymerization, it was effective as an inhibitor of arylsulfatase. This novel combretastatin A-4 sulfamate derivative has the potential to be developed as a dual inhibitor of tubulin polymerization and arylsulfatase for cancer therapy.
Collapse
Affiliation(s)
- Leilei Huang
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
| | - Jinwen Huang
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
- Shanghai Engineering Research Center of Green Fluoropharmaceutical Technology China
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry CAS China
| | - Hui Nie
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
| | - Yingzi Li
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
| | - Lixing Song
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
| | - Fanhong Wu
- Department of Pharmaceutical Engineering, School of Chemical and Environmental Engineering, Shanghai Institute of Technology Shanghai China
- Shanghai Engineering Research Center of Green Fluoropharmaceutical Technology China
| |
Collapse
|
10
|
Foster PA. Steroid Sulphatase and Its Inhibitors: Past, Present, and Future. Molecules 2021; 26:2852. [PMID: 34064842 PMCID: PMC8151039 DOI: 10.3390/molecules26102852] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/22/2022] Open
Abstract
Steroid sulphatase (STS), involved in the hydrolysis of steroid sulphates, plays an important role in the formation of both active oestrogens and androgens. Since these steroids significantly impact the proliferation of both oestrogen- and androgen-dependent cancers, many research groups over the past 30 years have designed and developed STS inhibitors. One of the main contributors to this field has been Prof. Barry Potter, previously at the University of Bath and now at the University of Oxford. Upon Prof. Potter's imminent retirement, this review takes a look back at the work on STS inhibitors and their contribution to our understanding of sulphate biology and as potential therapeutic agents in hormone-dependent disease. A number of potent STS inhibitors have now been developed, one of which, Irosustat (STX64, 667Coumate, BN83495), remains the only one to have completed phase I/II clinical trials against numerous indications (breast, prostate, endometrial). These studies have provided new insights into the origins of androgens and oestrogens in women and men. In addition to the therapeutic role of STS inhibition in breast and prostate cancer, there is now good evidence to suggest they may also provide benefits in patients with colorectal and ovarian cancer, and in treating endometriosis. To explore the potential of STS inhibitors further, a number of second- and third-generation inhibitors have been developed, together with single molecules that possess aromatase-STS inhibitory properties. The further development of potent STS inhibitors will allow their potential therapeutic value to be explored in a variety of hormone-dependent cancers and possibly other non-oncological conditions.
Collapse
Affiliation(s)
- Paul A. Foster
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK; ; Tel.: +44-121-414-3776
- Centre for Endocrinology, Metabolism and Diabetes, University of Birmingham, Birmingham Health Partners, Birmingham B15 2TT, UK
| |
Collapse
|
11
|
Jójárt R, Laczkó-Rigó R, Klement M, Kőhl G, Kecskeméti G, Özvegy-Laczka C, Mernyák E. Design, synthesis and biological evaluation of novel estrone phosphonates as high affinity organic anion-transporting polypeptide 2B1 (OATP2B1) inhibitors. Bioorg Chem 2021; 112:104914. [PMID: 33932771 DOI: 10.1016/j.bioorg.2021.104914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/07/2021] [Accepted: 04/11/2021] [Indexed: 12/12/2022]
Abstract
Organic anion-transporting polypeptide 2B1 (OATP2B1) is a multispecific membrane transporter mediating the cellular uptake of various exo- and endobiotics, including drugs and steroid hormones. Increased uptake of steroid hormones by OATP2B1 may increase tumor proliferation. Therefore, understanding OATP2B1's substrate/inhibitor recognition and inhibition of its function, e.g., in hormone-dependent tumors, would be highly desirable. To identify the crucial structural features that correlate with OATP2B1 inhibition, here we designed modifications at four positions of the estrane skeleton. 13α- or 13β-estrone phosphonates modified at ring A or ring D were synthesized. Hirao and Cu(I)-catalyzed azide-alkyne click reactions served in the syntheses as key steps. 13β-Derivatives displayed outstanding OATP2B1 inhibitory action with IC50 values in the nanomolar range (41-87 nM). A BODIPY-13α-estrone conjugate was additionally synthesized, modified at C-3-O of the steroid, containing a four-carbon linker between the triazole moiety and the BODIPY core. The fluorescent conjugate displayed efficient, submicromolar OATP2B1 inhibitory potency. The newly identified inhibitors and the structure-activity relationships specified here promote our understanding about drug recognition of OATP2B1.
Collapse
Affiliation(s)
- Rebeka Jójárt
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Réka Laczkó-Rigó
- Drug Resistance Research Group instead of Membrane Protein Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | - Máté Klement
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Gabriella Kőhl
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Gábor Kecskeméti
- Department of Medicinal Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Csilla Özvegy-Laczka
- Drug Resistance Research Group instead of Membrane Protein Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | - Erzsébet Mernyák
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary.
| |
Collapse
|
12
|
Kovács F, Gopisetty MK, Adamecz DI, Kiricsi M, Enyedy ÉA, Frank É. Synthesis and conversion of primary and secondary 2-aminoestradiols into A-ring-integrated benzoxazolone hybrids and their in vitro anticancer activity. RSC Adv 2021; 11:13885-13896. [PMID: 35423928 PMCID: PMC8697693 DOI: 10.1039/d1ra01889b] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 04/05/2021] [Indexed: 01/19/2023] Open
Abstract
Hybrid systems are often endowed with completely different and improved properties compared to their parent compounds. In order to extend the chemical space toward sterane-based molecular hybrids, a number of estradiol-derived benzoxazol-2-ones with combined aromatic rings were synthesized via the corresponding 2-aminophenol intermediates. 2-Aminoestradiol was first prepared from estrone by a two-step nitration/reduction sequence under mild reaction conditions. Subsequent reductive aminations with different arylaldehydes furnished secondary 2-aminoestradiol derivatives in good yields. The proton dissociation processes of the aminoestradiols were investigated in aqueous solution by UV-visible spectrophotometric titrations to reveal their actual chemical forms at physiological pH. The determined pK1 and pK2 values are attributed to the +NH3 or +NH2R and OH moieties, and both varied by the different R substituents of the amino group. Primary and secondary 2-aminoestradiols were next reacted with carbonyldiimidazole as a phosgene equivalent to introduce a carbonyl group with simultaneous ring-closure to give A-ring-fused oxazolone derivatives in high yields. The novel aminoestradiols and benzoxazolones were subjected to in vitro cytotoxicity analysis and were found to exert cancer cell specific activity. Estradiol–benzoxazolone hybrids with a common aromatic moiety were efficiently synthesized via primary and secondary aminophenol intermediates, and their anticancer activities were investigated.![]()
Collapse
Affiliation(s)
- Ferenc Kovács
- Department of Organic Chemistry, University of Szeged Dóm tér 8 H-6720 Szeged Hungary +36-62-544-275
| | - Mohana K Gopisetty
- Department of Biochemistry and Molecular Biology, Doctoral School of Biology, University of Szeged Közép fasor 52 H-6726 Szeged Hungary
| | - Dóra I Adamecz
- Department of Biochemistry and Molecular Biology, Doctoral School of Biology, University of Szeged Közép fasor 52 H-6726 Szeged Hungary
| | - Mónika Kiricsi
- Department of Biochemistry and Molecular Biology, Doctoral School of Biology, University of Szeged Közép fasor 52 H-6726 Szeged Hungary
| | - Éva A Enyedy
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged Dóm tér 7 H-6720 Szeged Hungary.,MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged Dóm tér 7 H-6720 Szeged Hungary
| | - Éva Frank
- Department of Organic Chemistry, University of Szeged Dóm tér 8 H-6720 Szeged Hungary +36-62-544-275
| |
Collapse
|
13
|
Daśko M, Demkowicz S, Biernacki K, Ciupak O, Kozak W, Masłyk M, Rachon J. Recent progress in the development of steroid sulphatase inhibitors - examples of the novel and most promising compounds from the last decade. J Enzyme Inhib Med Chem 2020; 35:1163-1184. [PMID: 32363947 PMCID: PMC7241464 DOI: 10.1080/14756366.2020.1758692] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/16/2020] [Accepted: 04/16/2020] [Indexed: 01/08/2023] Open
Abstract
The purpose of this review article is to provide an overview of recent achievements in the synthesis of novel steroid sulphatase (STS) inhibitors. STS is a crucial enzyme in the biosynthesis of active hormones (including oestrogens and androgens) and, therefore, represents an extremely attractive molecular target for the development of hormone-dependent cancer therapies. The inhibition of STS may effectively reduce the availability of active hormones for cancer cells, causing a positive therapeutic effect. Herein, we report examples of novel STS inhibitors based on steroidal and nonsteroidal cores that contain various functional groups (e.g. sulphamate and phosphorus moieties) and halogen atoms, which may potentially be used in therapies for hormone-dependent cancers. The presented work also includes examples of multitargeting agents with STS inhibitory activities. Furthermore, the fundamental discoveries in the development of the most promising drug candidates exhibiting STS inhibitory activities are highlighted.
Collapse
Affiliation(s)
- Mateusz Daśko
- Department of Inorganic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Sebastian Demkowicz
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Karol Biernacki
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Olga Ciupak
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Witold Kozak
- Department of Physical Chemistry, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Maciej Masłyk
- Department of Molecular Biology, Faculty of Biotechnology and Environment Sciences, The John Paul II Catholic University of Lublin, Lublin, Poland
| | - Janusz Rachon
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| |
Collapse
|
14
|
Jójárt R, Ali H, Horváth G, Kele Z, Zupkó I, Mernyák E. Pd-catalyzed Suzuki-Miyaura couplings and evaluation of 13α-estrone derivatives as potential anticancer agents. Steroids 2020; 164:108731. [PMID: 32946911 DOI: 10.1016/j.steroids.2020.108731] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/03/2020] [Accepted: 09/11/2020] [Indexed: 01/19/2023]
Abstract
13α-Estrones are of great value owing to their potent multiple bioactivity, including anticancer activity. 3-OH or 3-OBn derivatives of 2- or 4-[(subst.) phenyl]-13α-estrone as potential antiproliferative agents have been synthesized via facile, microwave-induced, Pd-catalyzed Suzuki-Miyaura coupling. 2- or 4-Halogenated 13α-estrone derivatives have been reacted with (4-subst.)phenylboronic acids using Pd(PPh3)4 as catalyst. The nature of para substituents at the introduced phenyl group did not influence the outcome of couplings. Certain newly synthesized compounds displayed substantial antiproliferative action against human adherent cancer cell lines of gynecological origin. Important structure-activity relationships were revealed, which might be helpful in the design of potent and selective anticancer derivatives based on the hormonally inactive 13α-estrane core.
Collapse
Affiliation(s)
- Rebeka Jójárt
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Hazhmat Ali
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6., H-6720 Szeged, Hungary
| | - Gergely Horváth
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Zoltán Kele
- Department of Medicinal Chemistry, University of Szeged, Dóm tér 8, 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
|
15
|
El-Gamal MI, Zaraei SO, Foster PA, Anbar HS, El-Gamal R, El-Awady R, Potter BVL. A new series of aryl sulfamate derivatives: Design, synthesis, and biological evaluation. Bioorg Med Chem 2020; 28:115406. [PMID: 32173116 DOI: 10.1016/j.bmc.2020.115406] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/27/2020] [Accepted: 02/29/2020] [Indexed: 12/12/2022]
Abstract
Steroid sulfatase (STS) has recently emerged as a drug target for management of hormone-dependent malignancies. In the present study, a new series of twenty-one aryl amido-linked sulfamate derivatives 1a-u was designed and synthesized, based upon a cyclohexyl lead compound. All members were evaluated as STS inhibitors in a cell-free assay. Adamantyl derivatives 1h and 1p-r were the most active with more than 90% inhibition at 10 µM concentration and, for those with the greatest inhibitory activity, IC50 values were determined. These compounds exhibited STS inhibition within the range of ca 25-110 nM. Amongst them, compound 1q possessing a o-chlorobenzene sulfamate moiety exhibited the most potent STS inhibitory activity with an IC50 of 26 nM. Furthermore, to assure capability to pass through the cell lipid bilayer, compounds with low IC50 values were tested against STS activity in JEG-3 whole-cell assays. Consequently, 1h and 1q demonstrated IC50 values of ca 14 and 150 nM, respectively. Thus, compound 1h is 31 times more potent than the corresponding cyclohexyl lead (IC50 value = 421 nM in a JEG-3 whole-cell assay). Furthermore, the most potent STS inhibitors (1h and 1p-r) were evaluated for their antiproliferative activity against the estrogen-dependent breast cancer cell line T-47D. They showed promising activity with single digit micromolar IC50 values (ca 1-6 µM) and their potency against T-47D cells was comparable to that against STS enzyme. In conclusion, this new class of adamantyl-containing aryl sulfamate inhibitor has potential for further development against hormone-dependent tumours.
Collapse
Affiliation(s)
- Mohammed I El-Gamal
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Medicinal Chemistry, Faculty of Pharmacy, University of Mansoura, Mansoura 35516, Egypt.
| | - Seyed-Omar Zaraei
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Paul A Foster
- Institute of Metabolism and Systems Research, 2nd Floor IBR Tower, University of Birmingham, Birmingham B15 2TT, United Kingdom; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, United Kingdom.
| | - Hanan S Anbar
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai 19099, United Arab Emirates
| | - Randa El-Gamal
- Department of Medical Biochemistry, Faculty of Medicine, University of Mansoura, Mansoura 35516, Egypt
| | - Raafat El-Awady
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Barry V L Potter
- Medicinal Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| |
Collapse
|
16
|
Sulfamates in drug design and discovery: Pre-clinical and clinical investigations. Eur J Med Chem 2019; 179:257-271. [PMID: 31255926 DOI: 10.1016/j.ejmech.2019.06.052] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 12/15/2022]
Abstract
In the present article, we reviewed the sulfamate-containing compounds reported as bioactive molecules. The possible molecular targets of sulfamate derivatives include steroid sulfatase enzyme, carbonic anhydrases, acyl transferase, and others. Sulfamate derivatives can help treat hormone-dependent tumors including breast, prostate, and endometrial cancers, Binge eating disorder, migraine, glaucoma, weight loss, and epilepsy. Sulfamate derivatives can act also as calcium sensing receptor agonists and can aid in osteoporosis. Furthermore, acyl sulfamate derivatives can act as antibacterial agents against Gram-positive bacteria. A recent study revealed a new side effect of topiramate, a sulfamate-containing compound, which is sialolithiasis. The structural and biological characteristics of the reviewed compounds are presented in detail.
Collapse
|
17
|
Saha T, Makar S, Swetha R, Gutti G, Singh SK. Estrogen signaling: An emanating therapeutic target for breast cancer treatment. Eur J Med Chem 2019; 177:116-143. [PMID: 31129450 DOI: 10.1016/j.ejmech.2019.05.023] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/15/2022]
Abstract
Breast cancer, a most common malignancy in women, was known to be associated with steroid hormone estrogen. The discovery of estrogen receptor (ER) gave us not only a powerful predictive and prognostic marker, but also an efficient target for the treatment of hormone-dependent breast cancer with various estrogen ligands. ER consists of two subtypes i.e. ERα and ERβ, that are mostly G-protein-coupled receptors and activated by estrogen, specially 17β-estradiol. The activation is followed by translocation into the nucleus and binding with DNA to modulate activities of different genes. ERs can manage synthesis of RNA through genomic actions without directly binding to DNA. Receptors are tethered by protein-protein interactions to a transcription factor complex to communicate with DNA. Estrogens also exhibit nongenomic actions, a characteristic feature of steroid hormones, which are so rapid to be considered by the activation of RNA and translation. These are habitually related to stimulation of different protein kinase cascades. Majority of post-menopausal breast cancer is estrogen dependent, mostly potent biological estrogen (E2) for continuous growth and proliferation. Estrogen helps in regulating the differentiation and proliferation of normal breast epithelial cells. In this review we have investigated the important role of ER in development and progression of breast cancer, which is complicated by receptor's interaction with co-regulatory proteins, cross-talk with other signal transduction pathways and development of treatment strategies viz. selective estrogen receptor modulators (SERMs), selective estrogen receptor down regulators (SERDs), aromatase and sulphatase inhibitors.
Collapse
Affiliation(s)
- Tanmay Saha
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India
| | - Subhajit Makar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India
| | - Rayala Swetha
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India
| | - Gopichand Gutti
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India
| | - Sushil K Singh
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, U.P, India.
| |
Collapse
|
18
|
Mohareb RM, Samir EM, Halim PA. Synthesis, and anti-proliferative, Pim-1 kinase inhibitors and molecular docking of thiophenes derived from estrone. Bioorg Chem 2018; 83:402-413. [PMID: 30415021 DOI: 10.1016/j.bioorg.2018.10.067] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 10/24/2018] [Accepted: 10/29/2018] [Indexed: 12/11/2022]
Abstract
Heterocyclization of steroids were reported to give biologically active products where ring D modification occured. Estrone (1) was used as a template to develop new heterocyclic compounds. Ring D modification of 1 through its reaction with cyanoacetylhydrazine and elemental sulfur gave the thiophene derivative 3. The latter compound reacted with acetophenone derivatives 4a-c to give the hydrazide-hydrazone derivatives 5a-c, respectively. In addition, compound 3 formed thiazole derivatives through its first reaction with phenylisothiocyanate to give the thiourea derivative 9 followed by the reaction of the later with α-halocarbonyl compounds. In the present work a series of novel estrone derivatives were designed, synthesized and evaluated for their in vitro biological activities against c-Met kinase, and six typical cancer cell lines (A549, H460, HT-29, MKN-45, U87MG and SMMC-7721). The most promising compounds 5b, 5c, 11a, 13c, 15b, 15c, 15d, 17a and 17b were further investigated against the five tyrosine kinases c-Kit, Flt-3, VEGFR-2, EGFR, and PDGFR. Compounds 5b, 15d, 17a and 17b were selected to examine their Pim-1 kinase inhibition activity where compounds 15d and 17b showed high activities. Molecular docking of some of the most potent compounds was demonstrated.
Collapse
Affiliation(s)
- Rafat M Mohareb
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt.
| | - Eman M Samir
- National Organization for Drug Control & Research, P.O. 29, Cairo, Egypt
| | - Peter A Halim
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
19
|
Ganeshapillai D, Woo LWL, Thomas MP, Purohit A, Potter BVL. C-3- and C-4-Substituted Bicyclic Coumarin Sulfamates as Potent Steroid Sulfatase Inhibitors. ACS OMEGA 2018; 3:10748-10772. [PMID: 30320251 PMCID: PMC6173509 DOI: 10.1021/acsomega.8b01383] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/16/2018] [Indexed: 06/08/2023]
Abstract
Synthetic routes to potent bicyclic nonsteroidal sulfamate-based active-site-directed inhibitors of the enzyme steroid sulfatase (STS), an emerging target in the treatment of postmenopausal hormone-dependent diseases, including breast cancer, are described. Sulfamate analogs 9-27 and 28-46 of the core in vivo active two-ring coumarin template, modified at the 4- and 3-positions, respectively, were synthesized to expand structure-activity relationships. α-Alkylacetoacetates were used to synthesize coumarin sulfamate derivatives with 3-position modifications, and the bicyclic ring of other parent coumarins was primarily constructed via the Pechmann synthesis of hydroxyl coumarins. Compounds were examined for STS inhibition in intact MCF-7 breast cancer cells and in placental microsomes. Low nanomolar potency STS inhibitors were achieved, and some were found to inhibit the enzyme in MCF-7 cells ca. 100-500 more potently than the parent 4-methylcoumarin-7-O-sulfamate 3, with the best compounds close in potency to the tricyclic clinical drug Irosustat. 3-Hexyl-4-methylcoumarin-7-O-sulfamate 29 and 3-benzyl-4-methylcoumarin-7-O-sulfamate 41 were particularly effective inhibitors with IC50 values of 0.68 and 1 nM in intact MCF-7 cells and 8 and 32 nM for placental microsomal STS, respectively. They were docked into the STS active site for comparison with estrone 3-O-sulfamate and Irosustat, showing their sulfamate group close to the catalytic hydrated formylglycine residue and their pendant group lying between the hydrophobic sidechains of L103, F178, and F488. Such highly potent STS inhibitors expand the structure-activity relationship for these coumarin sulfamate-based agents that possess therapeutic potential and may be worthy of further development.
Collapse
Affiliation(s)
- Dharshini Ganeshapillai
- Medicinal
Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2
7AY, U.K.
| | - L. W. Lawrence Woo
- Medicinal
Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2
7AY, U.K.
| | - Mark P. Thomas
- Medicinal
Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2
7AY, U.K.
| | - Atul Purohit
- Section
of Investigative Medicine, Diabetes, Endocrinology & Metabolism, Imperial College London, 6th Floor, Commonwealth Building (6N2B), Hammersmith
Hospital, Du Cane Road, London W12 0NN, U.K.
| | - Barry V. L. Potter
- Medicinal
Chemistry & Drug Discovery, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
- Medicinal
Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2
7AY, U.K.
| |
Collapse
|
20
|
Bacsa I, Szemerédi D, Wölfling J, Schneider G, Fekete L, Mernyák E. The first Pd-catalyzed Buchwald-Hartwig aminations at C-2 or C-4 in the estrone series. Beilstein J Org Chem 2018; 14:998-1003. [PMID: 29977371 PMCID: PMC6009172 DOI: 10.3762/bjoc.14.85] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/13/2018] [Indexed: 01/26/2023] Open
Abstract
A facile Pd-catalyzed C(sp2)–N coupling to provide a range of 2- or 4-[(subst.)phenyl]amino-13α-estrone derivatives has been achieved under microwave irradiation. The reactions were mediated with the use of Pd(OAc)2 as a catalyst and KOt-Bu as a base in the presence of X-Phos as a ligand. The desired products have been obtained in good to excellent yields. The nature and the position of the aniline substituent at the aromatic ring influenced the outcome of the couplings. 2-Amino-13α-estrone was also synthesized in a two-step protocol including an amination of 2-bromo-13α-estrone 3-benzyl ether with benzophenone imine and subsequent hydrogenolysis.
Collapse
Affiliation(s)
- Ildikó Bacsa
- Department of Organic Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary
| | - Dávid Szemerédi
- 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
| | - Lilla Fekete
- 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
| |
Collapse
|
21
|
Das D, Poddar P, Maity S, Samanta R. Rhodium(III)-Catalyzed C6-Selective Arylation of 2-Pyridones and Related Heterocycles Using Quinone Diazides: Syntheses of Heteroarylated Phenols. J Org Chem 2017; 82:3612-3621. [DOI: 10.1021/acs.joc.7b00135] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Debapratim Das
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Puja Poddar
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Saurabh Maity
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Rajarshi Samanta
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| |
Collapse
|
22
|
Harrelson JP, Lee MW. Expanding the view of breast cancer metabolism: Promising molecular targets and therapeutic opportunities. Pharmacol Ther 2016; 167:60-73. [DOI: 10.1016/j.pharmthera.2016.07.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/21/2016] [Indexed: 12/23/2022]
|
23
|
Kuriyama M, Hamaguchi N, Yano G, Tsukuda K, Sato K, Onomura O. Deuterodechlorination of Aryl/Heteroaryl Chlorides Catalyzed by a Palladium/Unsymmetrical NHC System. J Org Chem 2016; 81:8934-8946. [PMID: 27641511 DOI: 10.1021/acs.joc.6b01609] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The catalytic deuterodechlorination of aryl/heteroaryl chlorides was developed with a palladium/unsymmetrical NHC system, and the precisely controlled introduction of deuterium into a variety of aryl/heteroaryl compounds was achieved with a high level of efficiency, selectivity, and deuteration degree. This method was also successfully applied to the transformation of bioactive agents even in a gram-scale synthesis. The crystal structure analysis of Pd-NHC complexes led to the observation of Pd-arene interaction.
Collapse
Affiliation(s)
- Masami Kuriyama
- Graduate School of Biomedical Sciences, Nagasaki University , 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Norihisa Hamaguchi
- Graduate School of Biomedical Sciences, Nagasaki University , 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Gemba Yano
- Graduate School of Biomedical Sciences, Nagasaki University , 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Kotaro Tsukuda
- Graduate School of Biomedical Sciences, Nagasaki University , 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Kanako Sato
- Graduate School of Biomedical Sciences, Nagasaki University , 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Osamu Onomura
- Graduate School of Biomedical Sciences, Nagasaki University , 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| |
Collapse
|
24
|
El-Gamal MI, Semreen MH, Foster PA, Potter BVL. Design, synthesis, and biological evaluation of new arylamide derivatives possessing sulfonate or sulfamate moieties as steroid sulfatase enzyme inhibitors. Bioorg Med Chem 2016; 24:2762-7. [PMID: 27143133 PMCID: PMC4896991 DOI: 10.1016/j.bmc.2016.04.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/20/2016] [Accepted: 04/21/2016] [Indexed: 01/15/2023]
Abstract
A series of new arylamide derivatives possessing terminal sulfonate or sulfamate moieties was designed and synthesized. The target compounds were tested for in vitro inhibitory effects against the steroid sulfatase (STS) enzyme in a cell-free assay system. The free sulfamate derivative 1j was the most active. It inhibited the enzymatic activity by 72.0% and 55.7% at 20 μM and 10 μM, respectively. Compound 1j was further tested for STS inhibition in JEG-3 placental carcinoma cells with high STS enzyme activity. It inhibited 93.9% of the enzyme activity in JEG-3 placental carcinoma cells at 20 μM with an efficacy near to that of the well-established drug STX64 as reference. At 10 μM, 1j inhibited 86.1% of the STS activity of JEG-3. Its IC50 value against the STS enzyme in JEG-3 cells was 0.421 μM. Thus, 1j represents an attractive new non-steroidal lead for further optimization.
Collapse
Affiliation(s)
- Mohammed I El-Gamal
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Medicinal Chemistry, Faculty of Pharmacy, University of Mansoura, Mansoura 35516, Egypt.
| | - Mohammad H Semreen
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Paul A Foster
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2HT, United Kingdom.
| | - Barry V L Potter
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| |
Collapse
|
25
|
Ivanov A, Boldt S, Nisa ZU, Ali Shah SJ, Ehlers P, Villinger A, Schneider G, Wölfling J, Rahman Q, Iqbal J, Langer P. Synthesis and phosphatase inhibitory activity of 3-alkynylestrones and their derivatives. RSC Adv 2016. [DOI: 10.1039/c5ra25558a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
A range of 3-alkynylated 3-deoxy-estrones were prepared by Sonogashira reactions and transformed into estrone derived diones and quinoxalines.
Collapse
Affiliation(s)
- Anton Ivanov
- Institut für Chemie
- Universität Rostock
- 18059 Rostock
- Germany
| | | | - Zaib un Nisa
- Centre for Advanced Drug Research
- COMSATS Institute of Information Technology
- Abbottabad
- Pakistan
| | - Syed Jawad Ali Shah
- Centre for Advanced Drug Research
- COMSATS Institute of Information Technology
- Abbottabad
- Pakistan
| | - Peter Ehlers
- Institut für Chemie
- Universität Rostock
- 18059 Rostock
- Germany
- Leibniz-Institut für Katalyse an der Universität Rostock e.V
| | | | - Gyula Schneider
- Department of Organic Chemistry
- University of Szeged
- H-6720 Szeged
- Hungary
| | - János Wölfling
- Department of Organic Chemistry
- University of Szeged
- H-6720 Szeged
- Hungary
| | - Qamar Rahman
- Institut für Chemie
- Universität Rostock
- 18059 Rostock
- Germany
- Amity University
| | - Jamshed Iqbal
- Centre for Advanced Drug Research
- COMSATS Institute of Information Technology
- Abbottabad
- Pakistan
| | - Peter Langer
- Institut für Chemie
- Universität Rostock
- 18059 Rostock
- Germany
- Leibniz-Institut für Katalyse an der Universität Rostock e.V
| |
Collapse
|
26
|
Thomas MP, Potter BVL. Discovery and Development of the Aryl O-Sulfamate Pharmacophore for Oncology and Women's Health. J Med Chem 2015; 58:7634-58. [PMID: 25992880 PMCID: PMC5159624 DOI: 10.1021/acs.jmedchem.5b00386] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In 1994, following work from this laboratory, it was reported that estrone-3-O-sulfamate irreversibly inhibits a new potential hormone-dependent cancer target steroid sulfatase (STS). Subsequent drug discovery projects were initiated to develop the core aryl O-sulfamate pharmacophore that, over some 20 years, have led to steroidal and nonsteroidal drugs in numerous preclinical and clinical trials, with promising results in oncology and women's health, including endometriosis. Drugs have been designed to inhibit STS, e.g., Irosustat, as innovative dual-targeting aromatase-steroid sulfatase inhibitors (DASIs) and as multitargeting agents for hormone-independent tumors, such as the steroidal STX140 and nonsteroidal counterparts, acting inter alia through microtubule disruption. The aryl sulfamate pharmacophore is highly versatile, operating via three distinct mechanisms of action, and imbues attractive pharmaceutical properties. This Perspective gives a personal view of the work leading both to the therapeutic concepts and these drugs, their current status, and how they might develop in the future.
Collapse
Affiliation(s)
- Mark P. Thomas
- Wolfson Laboratory of Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom
| | - Barry V. L. Potter
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom
| |
Collapse
|
27
|
Mueller JW, Gilligan LC, Idkowiak J, Arlt W, Foster PA. The Regulation of Steroid Action by Sulfation and Desulfation. Endocr Rev 2015; 36:526-63. [PMID: 26213785 PMCID: PMC4591525 DOI: 10.1210/er.2015-1036] [Citation(s) in RCA: 285] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 07/21/2015] [Indexed: 12/14/2022]
Abstract
Steroid sulfation and desulfation are fundamental pathways vital for a functional vertebrate endocrine system. After biosynthesis, hydrophobic steroids are sulfated to expedite circulatory transit. Target cells express transmembrane organic anion-transporting polypeptides that facilitate cellular uptake of sulfated steroids. Once intracellular, sulfatases hydrolyze these steroid sulfate esters to their unconjugated, and usually active, forms. Because most steroids can be sulfated, including cholesterol, pregnenolone, dehydroepiandrosterone, and estrone, understanding the function, tissue distribution, and regulation of sulfation and desulfation processes provides significant insights into normal endocrine function. Not surprisingly, dysregulation of these pathways is associated with numerous pathologies, including steroid-dependent cancers, polycystic ovary syndrome, and X-linked ichthyosis. Here we provide a comprehensive examination of our current knowledge of endocrine-related sulfation and desulfation pathways. We describe the interplay between sulfatases and sulfotransferases, showing how their expression and regulation influences steroid action. Furthermore, we address the role that organic anion-transporting polypeptides play in regulating intracellular steroid concentrations and how their expression patterns influence many pathologies, especially cancer. Finally, the recent advances in pharmacologically targeting steroidogenic pathways will be examined.
Collapse
Affiliation(s)
- Jonathan W Mueller
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Lorna C Gilligan
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Jan Idkowiak
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Wiebke Arlt
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Paul A Foster
- Centre for Endocrinology, Diabetes, and Metabolism, Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
28
|
Thomas MP, Potter BVL. Estrogen O-sulfamates and their analogues: Clinical steroid sulfatase inhibitors with broad potential. J Steroid Biochem Mol Biol 2015; 153:160-9. [PMID: 25843211 DOI: 10.1016/j.jsbmb.2015.03.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/31/2015] [Indexed: 01/20/2023]
Abstract
Estrogen sulfamate derivatives were the first irreversible active-site-directed inhibitors of steroid sulfatase (STS), an emerging drug target for endocrine therapy of hormone dependent diseases that catalyzes inter alia the hydrolysis of estrone sulfate to estrone. In recent years this has stimulated clinical investigation of the estradiol derivative both as an oral prodrug and its currently ongoing exploration in endometriosis. 2-Substituted steroid sulfamate derivatives show considerable potential as multi-targeting agents for hormone-independent disease, but are also potent STS inhibitors. The steroidal template has spawned nonsteroidal STS inhibitors one of which, Irosustat, has been evaluated clinically in breast cancer, endometrial cancer and prostate cancer and there is potential for innovative dual-targeting approaches. This review surveys the role of estrogen sulfamates, their analogues and current status.
Collapse
Affiliation(s)
- Mark P Thomas
- Wolfson Laboratory of Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, United Kingdom
| | - Barry V L Potter
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom.
| |
Collapse
|
29
|
Abdoli M, Saeidian H. Synthesis and reactivity of imidazole-1-sulfonate esters (imidazylates) in substitution, elimination, and metal-catalyzed cross-coupling reactions: a review. J Sulphur Chem 2015. [DOI: 10.1080/17415993.2015.1057512] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
30
|
Yang B, Sun Z, Liu C, Cui Y, Guo Z, Ren Y, Lu Z, Knapp S. O-(Aminosulfonylation) of phenols and an example of slow hydrolytic release. Tetrahedron Lett 2014. [DOI: 10.1016/j.tetlet.2014.10.065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
31
|
(125)I-Labelled 2-Iodoestrone-3-sulfate: synthesis, characterization and OATP mediated transport studies in hormone dependent and independent breast cancer cells. Nucl Med Biol 2014; 42:274-82. [PMID: 25542669 DOI: 10.1016/j.nucmedbio.2014.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/28/2014] [Accepted: 10/29/2014] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Organic Anion Transporting Polypeptides (OATP) are a family of membrane associated transporters that facilitate estrone-3-sulphate (E3S) uptake by hormone dependent, post-menopausal breast cancers. We have established E3S as a potential ligand for targeting hormone dependent breast cancer cells, and in this study sought to prepare and investigate radioiodinated E3S as a tool to study the OATP system. METHODS 2- and 4-Iodoestrone-3-sulfates were prepared from estrone via aromatic iodination followed by a rapid and high yielding sulfation procedure. The resulting isomers were separated by preparative HPLC and verified by (1)H NMR and analytical HPLC. Transport studies of 2- and 4-[(125)I]-E3S were conducted in hormone dependent (i.e. MCF-7) and hormone independent (i.e. MDA-MB-231) breast cancer cells in the presence or absence of the specific transport inhibitor, bromosulfophthalein (BSP). Cellular localization of OATP1A2, OATP2B1, OATP3A1 and OATP4A1 were determined by immunofluorescence analysis using anti-Na(+)/K(+) ATPase-α (1:100 dilution) and DAPI as plasma membrane and nuclear markers, respectively. RESULTS Significantly (p<0.01) higher total accumulation of 2-[(125)I]-E3S was observed in hormone dependent MCF-7 as compared to hormone independent MDA-MB-231 breast cancer cells. In contrast 4-[(125)I]-E3S did not show cellular accumulation in either case. The efficiency of 2-[(125)I]-E3S transport (expressed as a ratio of Vmax/Km) was 2.4 times greater in the MCF-7 as compared to the MDA-MB-231 breast cancer cells. OATP1A2, OATP3A1 and OATP4A1 expression was localized in plasma membranes of MCF-7 and MDA-MB-231 cells confirming the functional role of these transporters in radioiodinated E3S cellular uptake. CONCLUSION An efficient method for the preparation of 2- and 4-[(125)I]-E3S was developed and where the former demonstrated potential as an in vitro probe for the OATP system. The new E3S probe can be used to study the OATP system and as a platform to create radiopharmaceuticals for imaging breast cancer.
Collapse
|
32
|
Majee D, Biswas S, Mobin SM, Samanta S. An expedient one-pot sequential three-component reaction for the stereoselective synthesis of functionalized spiro-sulfamidate imine fused δ-lactone scaffold. Tetrahedron Lett 2014. [DOI: 10.1016/j.tetlet.2014.06.088] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
33
|
Mohareb RM, Al-Omran F, Azzam RA. Heterocyclic ring extension of estrone: synthesis and cytotoxicity of fused pyran, pyrimidine and thiazole derivatives. Steroids 2014; 84:46-56. [PMID: 24686206 DOI: 10.1016/j.steroids.2014.03.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 02/21/2014] [Accepted: 03/15/2014] [Indexed: 01/03/2023]
Abstract
The one pot reaction of estrone with the aromatic aldehydes 2a-c and either of malononitrile or ethyl cyanoacetate afforded the fused pyran derivatives 4a-f. On the other hand, carrying the same reaction using thiourea instead of the cyanomethylene reagent gave the fused pyrimidine derivatives 6a-c. The latter compounds reacted with phenacyl bromide to give the thiazolo[3,2-a]pyrimidine derivatives 8a-c. The reaction of the title compound with bromine gave the monobromo derivative 13 which in turn reacted with either thiourea or cyanothioacetamide to give the thiazole derivatives 14 and 16, respectively. The cytotoxicity of the newly synthesized products was evaluated against six human cancer and normal cell lines where the results showed that compounds 4c, 4f, 6b, 8b, 8c, 10, 13, 16, 18c and 19c exhibited optimal cytotoxic effect against the cancer cell lines, with IC50's in the nM range.
Collapse
Affiliation(s)
- Rafat M Mohareb
- Department of Chemistry, Faculty of Science, Cairo University, Giza, A.R., Egypt.
| | - Fatima Al-Omran
- Department of Chemistry, Kuwait University, P.O. Box 5969, 13060 Safat, Kuwait
| | - Rasha A Azzam
- Department of Chemistry, Faculty of Science, Helwan University, Cairo, A.R., Egypt
| |
Collapse
|
34
|
Kajita D, Nakamura M, Matsumoto Y, Makishima M, Hashimoto Y. Design and synthesis of silicon-containing steroid sulfatase inhibitors possessing pro-estrogen antagonistic character. Bioorg Med Chem 2014; 22:2244-52. [PMID: 24630694 DOI: 10.1016/j.bmc.2014.02.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 02/10/2014] [Accepted: 02/10/2014] [Indexed: 11/26/2022]
Abstract
Steroid sulfatase (STS) is a potential target for treatment of postmenopausal hormone-dependent breast cancer. Several steroidal STS inhibitors have been reported, but steroidal compounds are difficult to optimize and may interact with other targets. On the other hand, we have shown that diphenylmethane (DPM) derivatives act as estrogen receptor (ER) agonists and antagonists. Here, we aimed to design and synthesize non-steroidal DPM-type STS inhibitors that would also serve as pro-estrogen antagonists, releasing a metabolite with ERα-antagonistic activity upon hydrolysis by STS. We synthesized a series of compounds and evaluated their biological activities by means of STS-inhibitory activity assay and ER reporter gene assay. Among them, silicon-containing compound 16a showed strong STS-inhibitory activity (IC50=0.17μM). Further, its putative metabolite (12a) exhibited potent ERα-antagonistic activity (IC50=29.7nM).
Collapse
Affiliation(s)
- Daisuke Kajita
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Masaharu Nakamura
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan.
| | - Yotaro Matsumoto
- Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Makoto Makishima
- Department of Biochemistry, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Yuichi Hashimoto
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| |
Collapse
|
35
|
Williams SJ, Denehy E, Krenske EH. Experimental and theoretical insights into the mechanisms of sulfate and sulfamate ester hydrolysis and the end products of type I sulfatase inactivation by aryl sulfamates. J Org Chem 2014; 79:1995-2005. [PMID: 24555731 DOI: 10.1021/jo4026513] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Type I sulfatases catalyze the hydrolysis of sulfate esters through S-O bond cleavage and possess a catalytically essential formylglycine (FGly) active-site residue that is post-translationally derived from either cysteine or serine. Type I sulfatases are inactivated by aryl sulfamates in a time-dependent, irreversible, and active-site directed manner consistent with covalent modification of the active site. We report a theoretical (SCS-MP2//B3LYP) and experimental study of the uncatalyzed and enzyme-catalyzed hydrolysis of aryl sulfates and sulfamates. In solution, aryl sulfate monoanions undergo hydrolysis by an S(N)2 mechanism whereas aryl sulfamate monoanions follow an S(N)1 pathway with SO2NH as an intermediate; theory traces this difference to the markedly greater stability of SO2NH versus SO3. For Pseudomonas aeruginosa arylsulfatase-catalyzed aryl sulfate hydrolysis, Brønsted analysis (log(V(max)/K(M)) versus leaving group pK(a) value) reveals β(LG) = -0.86 ± 0.23, consistent with an S(N)2 at sulfur reaction but substantially smaller than that reported for uncatalyzed hydrolysis (β(LG) = -1.81). Common to all proposed mechanisms of sulfatase catalysis is a sulfated FGly intermediate. Theory indicates a ≥26 kcal/mol preference for the intermediate to release HSO4(-) by an E2 mechanism, rather than alkaline phosphatase-like S(N)2 substitution by water. An evaluation of the stabilities of various proposed end-products of sulfamate-induced sulfatase inactivation highlights that an imine N-sulfate derived from FGly is the most likely irreversible adduct.
Collapse
Affiliation(s)
- Spencer J Williams
- School of Chemistry and ‡Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne , Melbourne, VIC 3010, Australia
| | | | | |
Collapse
|
36
|
Mostafa YA, Taylor SD. Steroid derivatives as inhibitors of steroid sulfatase. J Steroid Biochem Mol Biol 2013; 137:183-98. [PMID: 23391659 DOI: 10.1016/j.jsbmb.2013.01.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 01/10/2013] [Accepted: 01/25/2013] [Indexed: 10/27/2022]
Abstract
Sulfated steroids function as a storage reservoir of biologically active steroid hormones. The sulfated steroids themselves are biologically inactive and only become active in vivo when they are converted into their desulfated (unconjugated) form by the enzyme steroid sulfatase (STS). Inhibitors of STS are considered to be potential therapeutics for the treatment of steroid-dependent cancers such as breast, prostate and endometrial cancer. The present review summarizes steroid derivatives as inhibitors of STS covering the literature from the early years of STS inhibitor development to October of 2012. A brief discussion of the function, structure and mechanism of STS and its role in estrogen receptor-positive (ER+) hormone-dependent breast cancer is also presented. This article is part of a Special Issue entitled "Synthesis and biological testing of steroid derivatives as inhibitors".
Collapse
Affiliation(s)
- Yaser A Mostafa
- Department of Chemistry, University of Waterloo, 200 University Ave. West, Waterloo, ON, Canada
| | | |
Collapse
|
37
|
Woo LWL, Wood PM, Bubert C, Thomas MP, Purohit A, Potter BVL. Synthesis and structure-activity relationship studies of derivatives of the dual aromatase-sulfatase inhibitor 4-{[(4-cyanophenyl)(4H-1,2,4-triazol-4-yl)amino]methyl}phenyl sulfamate. ChemMedChem 2013; 8:779-99. [PMID: 23495205 PMCID: PMC3743159 DOI: 10.1002/cmdc.201300015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 02/13/2013] [Indexed: 02/04/2023]
Abstract
4-{[(4-Cyanophenyl)(4H-1,2,4-triazol-4-yl)amino]methyl}phenyl sulfamate and its ortho-halogenated (F, Cl, Br) derivatives are first-generation dual aromatase and sulfatase inhibitors (DASIs). Structure-activity relationship studies were performed on these compounds, and various modifications were made to their structures involving relocation of the halogen atom, introduction of more halogen atoms, replacement of the halogen with another group, replacement of the methylene linker with a difluoromethylene linker, replacement of the para-cyanophenyl ring with other ring structures, and replacement of the triazolyl group with an imidazolyl group. The most potent in vitro DASI discovered is an imidazole derivative with IC50 values against aromatase and steroid sulfatase in a JEG-3 cell preparation of 0.2 and 2.5 nM, respectively. The parent phenol of this compound inhibits aromatase with an IC50 value of 0.028 nM in the same assay.
Collapse
Affiliation(s)
- L W Lawrence Woo
- Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | | | | | | | | | | |
Collapse
|
38
|
Majee D, Srivastava A, Mobin SM, Samanta S. l-Proline catalyzed highly efficient synthesis of Z-5-alkylidene cyclic sulfamidate imines: an easy access to 5-alkyl-substituted cyclic sulfamidate imines. RSC Adv 2013. [DOI: 10.1039/c3ra40299a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
39
|
Abstract
INTRODUCTION Steroid sulfatase (STS) converts sulfated hormones to free hormones of importance in hormone-dependent diseases such as breast cancer and endometriosis. Carbohydrate sulfatases degrade complex carbohydrates as part of normal cellular turnover; certain lysosomal storage disorders (LSDs) involve defective processing of sulfated glycosaminoglycans by mutant sulfatases. AREAS COVERED Aryl sulfamates have been developed as STS inhibitors, and STX64 and PGL2001 are under evaluation in Phase I and II clinical trials for treatment of endometrial and metastatic breast and prostate cancers and endometriosis. Dual-acting compounds have emerged that are aromatase inhibitors (AIs), selective estrogen receptor antagonists, or inhibitors of microtubule polymerization. Sulfamidase inhibitors as pharmacological chaperones to assist maturation of folding-defective mutants for the treatment of Sanfilippo type A disease are under investigation. Coverage: The patent literature after the mid-1990s. EXPERT OPINION The failure of STX64 in a Phase II monotherapy clinical trial should not dissuade further investigations in multidrug regimens, particularly in combination with AIs. The recent development of dual-acting compounds may enhance the potential for success in the clinic. Further investigations into aryl sulfamates are required to clarify the molecular mechanism of action; additionally, new reversible sulfatase inhibition concepts are needed for the development of pharmacological chaperones for sulfatase LSDs.
Collapse
Affiliation(s)
- Spencer J Williams
- University of Melbourne, School of Chemistry and Bio21 Molecular Science, Parkville, Victoria, Australia.
| |
Collapse
|