1
|
Hosang L, Löhndorf A, Dohle W, Rosche A, Marry S, Diercks BP, Müller-Kirschbaum LC, Flügel LT, Potter BVL, Odoardi F, Guse AH, Flügel A. 2-Methoxyestradiol-3,17-O,O-bis-sulfamate inhibits store-operated Ca 2+ entry in T lymphocytes and prevents experimental autoimmune encephalomyelitis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119485. [PMID: 37150482 DOI: 10.1016/j.bbamcr.2023.119485] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/09/2023]
Abstract
Ca2+ signaling is one of the essential signaling systems for T lymphocyte activation, the latter being an essential step in the pathogenesis of autoimmune diseases such as multiple sclerosis (MS). Store-operated Ca2+ entry (SOCE) ensures long lasting Ca2+ signaling and is of utmost importance for major downstream T lymphocyte activation steps, e.g. nuclear localization of the transcription factor 'nuclear factor of activated T cells' (NFAT). 2-Methoxyestradiol (2ME2), an endogenous metabolite of estradiol (E2), blocks nuclear translocation of NFAT. The likely underlying mechanism is inhibition of SOCE, as shown for its synthetic sulfamate ester analogue 2-ethyl-3-sulfamoyloxy-17β-cyanomethylestra-1,3,5(10)-triene (STX564). Here, we demonstrate that another synthetic bis-sulfamoylated 2ME2 derivative, 2-methoxyestradiol-3,17-O,O-bis-sulfamate (2-MeOE2bisMATE, STX140), an orally bioavailable, multi-targeting anticancer agent and potent steroid sulfatase (STS) inhibitor, antagonized SOCE in T lymphocytes. Downstream events, e.g. secretion of the pro-inflammatory cytokines interferon-γ and interleukin-17, were decreased by STX140 in in vitro experiments. Remarkably, STX140 dosed in vivo completely blocked the clinical disease in both active and transfer experimental autoimmune encephalomyelitis (EAE) in Lewis rats, a T cell-mediated animal model for MS, at a dose of 10 mg/kg/day i.p., whereas neither 2ME2 nor Irosustat, a pure STS inhibitor, showed any effect. The STS inhibitory activity of STX140 is therefore not responsible for its activity in this model. Taken together, inhibition of SOCE by STX140 resulting in full antagonism of clinical symptoms in EAE in the Lewis rat, paired with the known excellent bioavailability and pharmaceutical profile of this drug, open potentially new therapeutic avenues for the treatment of MS.
Collapse
Affiliation(s)
- Leon Hosang
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Anke Löhndorf
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Wolfgang Dohle
- Drug Discovery & Medicinal Chemistry, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Anette Rosche
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Stephen Marry
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Björn-Philipp Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Lukas C Müller-Kirschbaum
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Lioba T Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany; Department of Neurology, University Medical Center Göttingen, Robert-Koch-Straße 40, D-37075 Göttingen, Germany
| | - Barry V L Potter
- Drug Discovery & Medicinal Chemistry, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Francesca Odoardi
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Andreas H Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany.
| |
Collapse
|
2
|
Mohamed A, Salah M, Tahoun M, Hawner M, Abdelsamie AS, Frotscher M. Dual Targeting of Steroid Sulfatase and 17β-Hydroxysteroid Dehydrogenase Type 1 by a Novel Drug-Prodrug Approach: A Potential Therapeutic Option for the Treatment of Endometriosis. J Med Chem 2022; 65:11726-11744. [PMID: 35993890 DOI: 10.1021/acs.jmedchem.2c00589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A novel approach for the dual inhibition of steroid sulfatase (STS) and 17β-hydroxysteroid dehydrogenase type 1(17β HSD1) by a single drug was explored, starting from in-house 17β HSD1 inhibitors via masking their phenolic OH group with a sulfamate ester. The sulfamates were intentionally designed as drugs for the inhibition of STS and, at the same time, prodrugs for 17β-HSD1 inhibition ("drug-prodrug approach"). The most promising sulfamates 13, 16, 18-20, 22-24, 36, and 37 showed nanomolar IC50 values for STS inhibition in a cellular assay and their corresponding phenols displayed potent 17β-HSD1 inhibition in cell-free and cellular assays, high selectivity over 17β-HSD2, reasonable metabolic stability, and low estrogen receptor α affinity. A close relationship was found between the liberation of the phenolic compound by sulfamate hydrolysis and 17β-HSD1 inactivation. These results showed that the envisaged drug-prodrug concept was successfully implemented. The novel compounds constitute a promising class of therapeutics for the treatment of endometriosis and other estrogen-dependent diseases.
Collapse
Affiliation(s)
- Abdelrahman Mohamed
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C23, Saarbrücken D-66123, Germany.,Pharmaceutical Organic Chemistry Department, Assiut University, Assiut 71526, Egypt
| | - Mohamed Salah
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C23, Saarbrücken D-66123, Germany.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, October University for Modern Sciences and Arts, Cairo 12451, Egypt
| | - Mariam Tahoun
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C23, Saarbrücken D-66123, Germany
| | - Manuel Hawner
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C23, Saarbrücken D-66123, Germany
| | - Ahmed S Abdelsamie
- Department of Chemistry of Natural and Microbial Products, Institute of Pharmaceutical and Drug Industries Research, National Research Centre, El-Buhouth St., Dokki, P.O. Box 12622 Cairo 12451, Egypt.,Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E81, Saarbrücken 66123, Germany
| | - Martin Frotscher
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C23, Saarbrücken D-66123, Germany
| |
Collapse
|
3
|
Saquib M, Baig MH, Khan MF, Azmi S, Khatoon S, Rawat AK, Dong JJ, Asad M, Arshad M, Hussain MK. Design and Synthesis of Bioinspired Benzocoumarin‐Chalcones Chimeras as Potential Anti‐Breast Cancer Agents. ChemistrySelect 2021. [DOI: 10.1002/slct.202101853] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Mohammad Saquib
- Department of Chemistry University of Allahabad Prayagraj (Allahabad) 211002, UP India
| | - Mohammad Hassan Baig
- Department of Family Medicine Gangnam Severance Hospital Yonsei University College of Medicine The Republic of Korea
| | - Mohammad Faheem Khan
- Department of Biotechnology Era's Lucknow Medical College Era University Lucknow 226003 UP India
| | - Sarfuddin Azmi
- Molecular Microbiology Biology Division Scientific Research Centre (SRC) Prince Sultan Military Medical City, Sulaimaniyah Riyadh 11159 Saudi Arabia
| | | | - Arun Kumar Rawat
- Department of Biochemistry Banaras Hindu University Varanasi 221005 UP India
| | - Jae June Dong
- Department of Family Medicine Gangnam Severance Hospital Yonsei University College of Medicine The Republic of Korea
| | - Mohammad Asad
- Chemistry Department Faculty of Science King Abdulaziz University, P.O. Box 80203 Jeddah 21589 Saudi Arabia
- Center of Excellence for Advanced Materials Research (CEAMR) King Abdulaziz University, P.O. Box 80203 Jeddah 21589 Saudi Arabia
| | - Md. Arshad
- Department of Zoology Aligarh Muslim University (AMU) Aligarh 202002 UP India
| | - Mohd Kamil Hussain
- Department of Chemistry Govt. Raza P.G. College Rampur M. J. P. Rohilkhand University 244901 Bareilly UP India
| |
Collapse
|
4
|
Ciupak O, Daśko M, Biernacki K, Rachon J, Masłyk M, Kubiński K, Martyna A, Demkowicz S. New potent steroid sulphatase inhibitors based on 6-(1-phenyl-1 H-1,2,3-triazol-4-yl)naphthalen-2-yl sulphamate derivatives. J Enzyme Inhib Med Chem 2021; 36:238-247. [PMID: 33322953 PMCID: PMC7744152 DOI: 10.1080/14756366.2020.1858820] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In the present work, we report a new class of potent steroid sulphatase (STS) inhibitors based on 6-(1-phenyl-1H-1,2,3-triazol-4-yl)naphthalen-2-yl sulphamate derivatives. Within the set of new STS inhibitors, 6-(1-(1,2,3-trifluorophenyl)-1H-1,2,3-triazol-4-yl)naphthalen-2-yl sulphamate 3L demonstrated the highest activity in the enzymatic assay inhibiting the STS activity to 7.98% at 0.5 µM concentration. Furthermore, to verify whether the obtained STS inhibitors are able to pass through the cellular membrane effectively, cell line experiments have been carried out. We found that the lowest STS activities were measured in the presence of compound 3L (remaining STS activity of 5.22%, 27.48% and 99.0% at 100, 10 and 1 nM concentrations, respectively). The measured STS activities for Irosustat (used as a reference) were 5.72%, 12.93% and 16.83% in the same concentration range. Moreover, a determined IC50 value of 15.97 nM for 3L showed that this compound is a very promising candidate for further preclinical investigations.
Collapse
Affiliation(s)
- Olga Ciupak
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Mateusz Daśko
- Department of Inorganic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Karol Biernacki
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Janusz Rachon
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Maciej Masłyk
- Department of Molecular Biology, The John Paul II Catholic University of Lublin, Lublin, Poland
| | - Konrad Kubiński
- Department of Molecular Biology, The John Paul II Catholic University of Lublin, Lublin, Poland
| | - Aleksandra Martyna
- Department of Molecular Biology, The John Paul II Catholic University of Lublin, Lublin, Poland
| | - Sebastian Demkowicz
- Department of Organic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Grienke U, Kaserer T, Kirchweger B, Lambrinidis G, Kandel RT, Foster PA, Schuster D, Mikros E, Rollinger JM. Steroid sulfatase inhibiting lanostane triterpenes - Structure activity relationship and in silico insights. Bioorg Chem 2019; 95:103495. [PMID: 31855822 DOI: 10.1016/j.bioorg.2019.103495] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/19/2019] [Accepted: 12/04/2019] [Indexed: 12/29/2022]
Abstract
Steroid sulfatase (STS) transforms hormone precursors into active steroids. Thus, it represents a target of intense research regarding hormone-dependent cancers. In this study, three ligand-based pharmacophore models were developed to identify STS inhibitors from natural sources. In a pharmacophore-based virtual screening of a curated molecular TCM database, lanostane-type triterpenes (LTTs) were predicted as STS ligands. Three traditionally used polypores rich in LTTs, i.e., Ganoderma lucidum Karst., Gloeophyllum odoratum Imazeki, and Fomitopsis pinicola Karst., were selected as starting materials. Based on eighteen thereof isolated LTTs a structure activity relationship for this compound class was established with piptolinic acid D (1), pinicolic acid B (2), and ganoderol A (3) being the most pronounced and first natural product STS inhibitors with IC50 values between 10 and 16 µM. Molecular docking studies proposed crucial ligand target interactions and a prediction tool for these natural compounds correlating with experimental findings.
Collapse
Affiliation(s)
- Ulrike Grienke
- Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Althanstraße 14, 1090 Vienna, Austria.
| | - Teresa Kaserer
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80-82, 6020 Innsbruck Austria
| | - Benjamin Kirchweger
- Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| | - George Lambrinidis
- Department of Pharmacy, Division of Pharmaceutical Chemistry, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - Ralph T Kandel
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80-82, 6020 Innsbruck Austria
| | - 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, United Kingdom
| | - Daniela Schuster
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80-82, 6020 Innsbruck Austria; Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Emmanuel Mikros
- Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Althanstraße 14, 1090 Vienna, Austria; Department of Pharmacy, Division of Pharmaceutical Chemistry, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - Judith M Rollinger
- Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| |
Collapse
|
8
|
Synthesis and in vitro evaluation of piperazinyl-ureido sulfamates as steroid sulfatase inhibitors. Eur J Med Chem 2019; 182:111614. [PMID: 31422224 DOI: 10.1016/j.ejmech.2019.111614] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/26/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022]
Abstract
Two new piperazinyl-ureido single ring aryl sulfamate-based inhibitor series were designed against the emerging oncology drug target steroid sulfatase (STS), for which there are existing potent steroidal and non-steroidal agents in clinical trials. 4-(Piperazinocarbonyl)aminosulfamates (5-31) were obtained by reacting 4-hydroxyarylamines with phenylchloroformate, subsequent sulfamoylation of the resulting hydroxyarylcarbamates and coupling of the product with 1-substituted piperazines. Pyrimidinyl-piperazinourea sulfamates (35-42) were synthesized by pyrimidine ring closure of 4-Boc-piperazine-1-carboxamidine with 3-(dimethylamino)propenones, deprotection and coupling with the sulfamoylated building block. Target ureidosulfamates 5-31 and 35-42 were evaluated both as STS inhibitors in vitro using a lysate of JEG-3 human placenta choriocarcinoma cell line and in a whole cell assay. SAR conclusions were drawn from both series. In series 35-42 the best inhibitory activity is related to the presence of a benzofuryl on the pyrimidine ring. In series 5-31 the best inhibitory activity was shown by the ureas bearing 4-chlorophenyl, 3,4-dichlorophenyl groups or aliphatic chains at the piperazino 4-nitrogen displaying IC50 in the 33-94 nM concentration range. Final optimization to the low nanomolar level was achieved through substitution of the arylsulfamate ring with halogens. Four halogenated arylsulfamates of high potency were achieved and two of these 19 and 20 had IC50 values of 5.1 and 8.8 nM respectively and are attractive for potential in vivo evaluation and further development. We demonstrate the optimization of this new series to low nanomolar potency, employing fluorine substitution, providing potent membrane permeant inhibitors with further development potential indicating piperazinyl-ureido aryl sulfamate derivatives as an attractive new class of STS inhibitors.
Collapse
|
9
|
Humby T, Davies W. Brain Gene Expression in a Novel Mouse Model of Postpartum Mood Disorder. Transl Neurosci 2019; 10:168-174. [PMID: 31410299 PMCID: PMC6689211 DOI: 10.1515/tnsci-2019-0030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 06/26/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Steroid sulfatase (STS) cleaves sulfate groups from steroid hormones; its expression/activity increases in late pregnancy and into the postpartum period. STS-deficient human and mouse mothers display elevated psychopathology and abnormal behaviour respectively; in mice, these effects can be partially normalised by antipsychotic (ziprasidone) administration. METHODOLOGY We compared brain gene expression in new mouse mothers administered the STS inhibitor 667-Coumate, or vehicle; significant changes were followed-up with pathway analysis and quantitative polymerase chain reaction (qPCR). Finally, the effects of combined 667-Coumate and ziprasidone administration on expression of the most robustly differentially-expressed genes were examined. RESULTS Surprisingly, no between-group gene expression changes were detected at a False Discovery Rate (FDR)-corrected p<0.1. 1,081 unique expression changes were detected at p<0.05, two top hits were verified by qPCR, and pathway analysis indicated enrichment of genes involved in olfactory transduction. The expression of Stoml3 and Cyp2g1 was unaffected by ziprasidone administration. CONCLUSIONS Postpartum behavioural abnormalities in STS-deficient mothers are likely to be the culmination of many small gene expression changes. Our data are consistent with the idea that olfactory function is key to maternal behaviour in mice, and suggest that aberrant expression of olfactory system genes may underlie abnormal maternal behaviour in STS-deficient women.
Collapse
Affiliation(s)
- Trevor Humby
- School of Psychology, Cardiff University, Cardiff CF10 3AT, Cardiff, UK
- Neuroscience and Mental Health Research Institute, Cardiff University, CardiffCF24 4HQ, UK
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics and Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, CardiffCF24 4HQ, UK
| | - William Davies
- School of Psychology, Cardiff University, Cardiff CF10 3AT, Cardiff, UK
- Neuroscience and Mental Health Research Institute, Cardiff University, CardiffCF24 4HQ, UK
- Medical Research Council Centre for Neuropsychiatric Genetics and Genomics and Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, CardiffCF24 4HQ, UK
| |
Collapse
|
10
|
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
|
11
|
Kaurav MS, Sahu PK, Sahu PK, Messali M, Almutairi SM, Sahu PL, Agarwal DD. An efficient, mild and metal free l-proline catalyzed construction of fused pyrimidines under microwave conditions in water. RSC Adv 2019; 9:3755-3763. [PMID: 35518091 PMCID: PMC9060310 DOI: 10.1039/c8ra07517d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 01/10/2019] [Indexed: 02/03/2023] Open
Abstract
One-pot condensation of 4-hydroxy coumarins, aldehydes and urea/thiourea to build C-C and C-N bonds is described. Fused pyrimidines have been synthesized under mild reaction conditions using l-proline. The protocol has been performed rapidly and efficiently in water under metal free conditions. Heterocyclic derivatives have been synthesized using the present methodology and avoid the use of hazardous solvents over conventional organic solvents. A proposed mechanism could be established for three component reactions. The present study reveals the first case in which l-proline has been explored as a homogeneous catalyst in the synthesis of fused pyrimidines in water under microwave irradiation. This synthesis involves simple workup and acceptable efficiency. The most notable feature of this protocol is the ability of the catalyst to influence asymmetric induction in the reaction.
Collapse
Affiliation(s)
- Manvendra S Kaurav
- School of Studies in Chemistry, Jiwaji University Gwalior-474011 Madhya Pradesh India
| | - Pramod K Sahu
- School of Studies in Chemistry, Jiwaji University Gwalior-474011 Madhya Pradesh India
- Department of Industrial Chemistry, Jiwaji University Gwalior-474011 Madhya Pradesh India
| | - Praveen K Sahu
- Department of Industrial Chemistry, Jiwaji University Gwalior-474011 Madhya Pradesh India
| | - Mouslim Messali
- Department of Chemistry, Taibah University 30002 Al-Madina Al-Mounawara Saudi Arabia
| | - Saud M Almutairi
- King Abdulaziz City for Science and Technology P. O. Box 6086 Riyadh 11442 Saudi Arabia
| | - Puran L Sahu
- Indian Pharmacopoeia Commission, Ministry of Health and Family Welfare Sector-23, Raj Nagar Ghaziabad 201002 India
- National Dope Testing Laboratory (NDTL), Ministry of Youth Affair & Sports, Government of India J. L. N. Stadium Complex East Gate No. 10, Lodi Road New Delhi-3 India
| | - Dau D Agarwal
- School of Studies in Chemistry, Jiwaji University Gwalior-474011 Madhya Pradesh India
- Department of Industrial Chemistry, Jiwaji University Gwalior-474011 Madhya Pradesh India
| |
Collapse
|
12
|
Kurogi K, Yoshihama M, Williams FE, Kenmochi N, Sakakibara Y, Suiko M, Liu MC. Identification of zebrafish steroid sulfatase and comparative analysis of the enzymatic properties with human steroid sulfatase. J Steroid Biochem Mol Biol 2019; 185:110-117. [PMID: 30118815 PMCID: PMC6289849 DOI: 10.1016/j.jsbmb.2018.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/11/2018] [Accepted: 08/08/2018] [Indexed: 11/19/2022]
Abstract
Steroid sulfatase (STS) plays an important role in the regulation of steroid hormones. Metabolism of steroid hormones in zebrafish has been investigated, but the action of steroid sulfatase remains unknown. In this study, a zebrafish sts was cloned, expressed, purified, and characterized in comparison with the orthologous human enzyme. Enzymatic assays demonstrated that similar to human STS, zebrafish Sts was most active in catalyzing the hydrolysis of estrone-sulfate and estradiol-sulfate, among five steroid sulfates tested as substrates. Kinetic analyses revealed that the Km values of zebrafish Sts and human STS differed with respective substrates, but the catalytic efficiency as reflected by the Vmax/Km appeared comparable, except for DHEA-sulfate with which zebrafish Sts appeared less efficient. While zebrafish Sts was catalytically active at 28 °C, the enzyme appeared more active at 37 °C and with similar Km values to those determined at 28 °C. Assays performed in the presence of different divalent cations showed that the activities of both zebrafish and human STSs were stimulated by Ca2+, Mg2+, and Mn2+, and inhibited by Zn+2 and Fe2+. EMATE and STX64, two known mammalian steroid sulafatase inhibitors, were shown to be capable of inhibiting the activity of zebrafish Sts. Collectively, the results obtained indicated that zebrafish Sts exhibited enzymatic characteristics comparable to the human STS, suggesting that the physiological function of STS may be conserved between zebrafish and humans.
Collapse
Affiliation(s)
- Katsuhisa Kurogi
- Department of Pharmacology, College of Pharmacy, University of Toledo Health Science Campus, Toledo, OH 43614, USA; Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki 889-2192, Japan
| | - Maki Yoshihama
- Department of Pharmacology, College of Pharmacy, University of Toledo Health Science Campus, Toledo, OH 43614, USA; Frontier Research Center, University of Miyazaki, Miyazaki 889-2192, Japan
| | - Frederick E Williams
- Department of Pharmacology, College of Pharmacy, University of Toledo Health Science Campus, Toledo, OH 43614, USA
| | - Naoya Kenmochi
- Frontier Research Center, University of Miyazaki, Miyazaki 889-2192, Japan
| | - Yoichi Sakakibara
- Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki 889-2192, Japan
| | - Masahito Suiko
- Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki 889-2192, Japan
| | - Ming-Cheh Liu
- Department of Pharmacology, College of Pharmacy, University of Toledo Health Science Campus, Toledo, OH 43614, USA.
| |
Collapse
|
13
|
Sahu PK, Sahu PK, Kaurav MS, Messali M, Almutairi SM, Sahu PL, Agarwal DD. Metal-Free Construction of Fused Pyrimidines via Consecutive C-C and C-N Bond Formation in Water. ACS OMEGA 2018; 3:15035-15042. [PMID: 31458170 PMCID: PMC6643823 DOI: 10.1021/acsomega.8b01993] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 10/10/2018] [Indexed: 05/22/2023]
Abstract
A facile and efficient protocol has been developed for mild construction of fused pyrimidines via l-proline-catalyzed reaction of 4-hydroxy coumarins, aldehydes, and 2-aminobenzothiazoles/urea. The reaction has been carried out rapidly and efficiently in water under mild and metal-free conditions. Current etiquette has efficiently synthesized the heterocycles and avoids the use of hazardous solvents over conventional organic solvents. A plausible reaction mechanism has been established in this study. This study represents the first case in which l-proline as a homogeneous catalyst has been explored in the synthesis of fused pyrimidines in water in view of simple procedure and acceptable efficiency. This method gives the target product in excellent yield with ease of workup.
Collapse
Affiliation(s)
- Pramod K. Sahu
- School of Studies in Chemistry and Department of Industrial Chemistry, Jiwaji University, Gwalior 474011, Madhya Pradesh, India
- E-mail: , (Pramod Kumar Sahu)
| | - Praveen K. Sahu
- School of Studies in Chemistry and Department of Industrial Chemistry, Jiwaji University, Gwalior 474011, Madhya Pradesh, India
| | - Manvendra S. Kaurav
- School of Studies in Chemistry and Department of Industrial Chemistry, Jiwaji University, Gwalior 474011, Madhya Pradesh, India
| | - Mouslim Messali
- Department
of Chemistry, Taibah University, 30002 Al-Madina
Al-Mounawara, Saudi Arabia
| | - Saud M. Almutairi
- King Abdulaziz
City for Science and Technology, P.O.
Box 6086, Riyadh 11442, Saudi Arabia
| | - Puran L. Sahu
- Indian Pharmacopoeia Commission Ministry
of Health and Family Welfare, Sector-23, Raj Nagar, Ghaziabad 201002, India
| | - Dau D. Agarwal
- School of Studies in Chemistry and Department of Industrial Chemistry, Jiwaji University, Gwalior 474011, Madhya Pradesh, India
| |
Collapse
|
14
|
Dhawan S, Kerru N, Awolade P, Singh-Pillay A, Saha ST, Kaur M, Jonnalagadda SB, Singh P. Synthesis, computational studies and antiproliferative activities of coumarin-tagged 1,3,4-oxadiazole conjugates against MDA-MB-231 and MCF-7 human breast cancer cells. Bioorg Med Chem 2018; 26:5612-5623. [PMID: 30360952 DOI: 10.1016/j.bmc.2018.10.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/02/2018] [Accepted: 10/10/2018] [Indexed: 11/16/2022]
Abstract
A novel library of coumarin tagged 1,3,4 oxadiazole conjugates was synthesized and evaluated for their antiproliferative activities against MDA-MB-231 and MCF-7 breast cancer cell lines. The evaluation studies revealed that compound 9d was the most potent molecule with an IC50 value of <5 µM against the MCF-7 cell line. Interestingly, compounds 10b and 11a showed a similar trend with lower inhibitory concentration (IC50 = 7.07 µM), in Estrogen Negative (ER-) cells than Estrogen Positive (ER+) cells. Structure-activity relationship (SAR) studies revealed that conjugates bearing benzyl moieties (9b, 9c and 9d) had superior activities compared to their alkyl analogues. The most potent compound 9d showed ∼1.4 times more potent activity than tamoxifen against MCF-7 cell line; while the introduction of sulfone unit in compounds 11a, 11b and 11c resulted in significant cytotoxicity against both MCF-7 and MDA-MB-231 cell lines. These results were further supported by docking studies, which revealed that the stronger binding affinity of the synthesized conjugates is due to the presence of sulfone unit attached to the substituted benzyl moiety in their pharmacophores.
Collapse
Affiliation(s)
- Sanjeev Dhawan
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban 4000, South Africa
| | - Nagaraju Kerru
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban 4000, South Africa
| | - Paul Awolade
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban 4000, South Africa
| | - Ashona Singh-Pillay
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban 4000, South Africa
| | - Sourav Taru Saha
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, Wits, Johannesburg 2050, South Africa
| | - Mandeep Kaur
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, Wits, Johannesburg 2050, South Africa
| | - Sreekantha B Jonnalagadda
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban 4000, South Africa
| | - Parvesh Singh
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban 4000, South Africa.
| |
Collapse
|
15
|
Sang X, Han H, Poirier D, Lin SX. Steroid sulfatase inhibition success and limitation in breast cancer clinical assays: An underlying mechanism. J Steroid Biochem Mol Biol 2018; 183:80-93. [PMID: 29803725 DOI: 10.1016/j.jsbmb.2018.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 04/18/2018] [Accepted: 05/23/2018] [Indexed: 12/23/2022]
Abstract
Steroid sulfatase is detectable in most hormone-dependent breast cancers. STX64, an STS inhibitor, induced tumor reduction in animal assay. Despite success in phase І clinical trial, the results of phase II trial were not that significant. Breast Cancer epithelial cells (MCF-7 and T47D) were treated with two STS inhibitors (STX64 and EM1913). Cell proliferation, cell cycle, and the concentrations of estradiol and 5α-dihydrotestosterone were measured to determine the endocrinological mechanism of sulfatase inhibition. Comparisons were made with inhibitions of reductive 17β-hydroxysteroid dehydrogenases (17β-HSDs). Proliferation studies showed that DNA synthesis in cancer cells was modestly decreased (approximately 20%), accompanied by an up to 6.5% in cells in the G0/G1 phase and cyclin D1 expression reduction. The concentrations of estradiol and 5α-dihydrotestosterone were decreased by 26% and 3% respectively. However, supplementation of 5α-dihydrotestosterone produced a significant increase (approximately 35.6%) in the anti-proliferative effect of sulfatase inhibition. This study has clarified sex-hormone control by sulfatase in BC, suggesting that the different roles of estradiol and 5α-dihydrotestosterone can lead to a reduction in the effect of sulfatase inhibition when compared with 17β-HSD7 inhibition. This suggests that combined treatment of sulfatase inhibitors with 17β-HSD inhibitors such as the type7 inhibitor could hold promise for hormone-dependent breast cancer.
Collapse
Affiliation(s)
- Xiaoye Sang
- Laboratory of Molecular Endocrinology and Oncology, CHU de Quebec-Research Center (CHUL) and Laval University, 2705 Boulevard Laurier, Québec City, Québec, G1V4G2, Canada
| | - Hui Han
- Laboratory of Molecular Endocrinology and Oncology, CHU de Quebec-Research Center (CHUL) and Laval University, 2705 Boulevard Laurier, Québec City, Québec, G1V4G2, Canada; Department of Thyroid Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Donald Poirier
- Laboratory of Molecular Endocrinology and Oncology, CHU de Quebec-Research Center (CHUL) and Laval University, 2705 Boulevard Laurier, Québec City, Québec, G1V4G2, Canada
| | - Sheng-Xiang Lin
- Laboratory of Molecular Endocrinology and Oncology, CHU de Quebec-Research Center (CHUL) and Laval University, 2705 Boulevard Laurier, Québec City, Québec, G1V4G2, Canada.
| |
Collapse
|
16
|
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
|
17
|
Potter BVL. SULFATION PATHWAYS: Steroid sulphatase inhibition via aryl sulphamates: clinical progress, mechanism and future prospects. J Mol Endocrinol 2018; 61:T233-T252. [PMID: 29618488 DOI: 10.1530/jme-18-0045] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 04/04/2018] [Indexed: 12/13/2022]
Abstract
Steroid sulphatase is an emerging drug target for the endocrine therapy of hormone-dependent diseases, catalysing oestrogen sulphate hydrolysis to oestrogen. Drug discovery, developing the core aryl O-sulphamate pharmacophore, has led to steroidal and non-steroidal drugs entering numerous clinical trials, with promising results in oncology and women's health. Steroidal oestrogen sulphamate derivatives were the first irreversible active-site-directed inhibitors and one was developed clinically as an oral oestradiol pro-drug and for endometriosis applications. This review summarizes work leading to the therapeutic concept of sulphatase inhibition, clinical trials executed to date and new insights into the mechanism of inhibition of steroid sulphatase. To date, the non-steroidal sulphatase inhibitor Irosustat has been evaluated clinically in breast cancer, alone and in combination, in endometrial cancer and in prostate cancer. The versatile core pharmacophore both imbues attractive pharmaceutical properties and functions via three distinct mechanisms of action, as a pro-drug, an enzyme active-site-modifying motif, likely through direct sulphamoyl group transfer, and as a structural component augmenting activity, for example by enhancing interactions at the colchicine binding site of tubulin. Preliminary new structural data on the Pseudomonas aeruginosa arylsulphatase enzyme suggest two possible sulphamate-based adducts with the active site formylglycine as candidates for the inhibition end product via sulphamoyl or sulphonylamine transfer, and a speculative choice is suggested. The clinical status of sulphatase inhibition is surveyed and how it might develop in the future. Also discussed are dual-targeting approaches, development of 2-substituted steroidal sulphamates and non-steroidal derivatives as multi-targeting agents for hormone-independent tumours, with other emerging directions.
Collapse
Affiliation(s)
- Barry V L Potter
- Medicinal Chemistry & Drug DiscoveryDepartment of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
18
|
Penning TM. Dehydroepiandrosterone (DHEA)-SO 4 Depot and Castration-Resistant Prostate Cancer. VITAMINS AND HORMONES 2018; 108:309-331. [PMID: 30029732 DOI: 10.1016/bs.vh.2018.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dehydroepiandrosterone (DHEA)-SO4 of adrenal origin is the major C19 steroid in the serum. It is a precursor of intratumoral androgen biosynthesis in patients with advanced prostate cancer following chemical or surgical castration. DHEA is a product of the P450c17 (17α-hydroxylase-17,20-lyase) enzyme. Despite inhibition of P450c17 with new agents, e.g., Abiraterone acetate, Orterenol, and Galeterone, the level of enzyme inhibition rarely exceeds 90% leaving behind a significant depot for androgen biosynthesis within the tumor. For DHEA-SO4 to be utilized there is uptake by organic anion transporter polypeptides, deconjugation catalyzed by steroid sulfatase, and adaptive upregulation of prostate steroidogenic enzymes that will convert DHEA into either testosterone or dihydrotestosterone. The depot of DHEA-SO4 that remains after P450c17 inhibition and the adaptive responses that occur within the tumor to promote DHEA utilization contribute to mechanisms of drug resistance observed with P450c17 inhibitors. Knowledge of these mechanisms identify new targets for therapeutics that could be used to surmount drug resistance in prostate cancer.
Collapse
Affiliation(s)
- Trevor M Penning
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
19
|
Iqbal J, El-Gamal MI, Ejaz SA, Lecka J, Sévigny J, Oh CH. Tricyclic coumarin sulphonate derivatives with alkaline phosphatase inhibitory effects: in vitro and docking studies. J Enzyme Inhib Med Chem 2018; 33:479-484. [PMID: 29390901 PMCID: PMC6009858 DOI: 10.1080/14756366.2018.1428193] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Tissue-nonspecific alkaline phosphatase (TNAP) is an important isozyme of alkaline phosphatases, which plays different pivotal roles within the human body. Most importantly, it is responsible for maintaining the balanced ratio of phosphate and inorganic pyrophosphate, thus regulates the extracellular matrix calcification during bone formation and growth. The elevated level of TNAP has been linked to vascular calcification and end-stage renal diseases. Consequently, there is a need to search for highly potent and selective inhibitors of alkaline phosphatases (APs) for treatment of disorders associated with the over-expression of APs. Herein, a series of tricyclic coumarin sulphonate 1a-za with known antiproliferative activity, was evaluated for AP inhibition against human tissue nonspecific alkaline phosphatase (h-TNAP) and human intestinal alkaline phosphatase (h-IAP). The methylbenzenesulphonate derivative 1f (IC50 = 0.38 ± 0.01 μM) was found to be the most active h-TNAP inhibitor. Another 4-fluorobenzenesulphonate derivative 1i (IC50 = 0.45 ± 0.02 μM) was found as the strongest inhibitor of h-IAP. Some of the derivatives were also identified as highly selective inhibitors of APs. Detailed structure-activity relationship (SAR) was investigated to identify the functional groups responsible for the effective inhibition of AP isozymes. The study was also supported by the docking studies to rationalise the most possible binding site interactions of the identified inhibitors with the targeted enzymes.
Collapse
Affiliation(s)
- Jamshed Iqbal
- a Centre for Advanced Drug Research , COMSATS Institute of Information Technology , Abbottabad , Pakistan
| | - Mohammed I El-Gamal
- b Department of Medicinal Chemistry, College of Pharmacy , University of Sharjah , Sharjah , United Arab Emirates.,c Sharjah Institute for Medical Research , University of Sharjah , Sharjah , United Arab Emirates.,d Department of Medicinal Chemistry , University of Mansoura , Mansoura , Egypt
| | - Syeda Abida Ejaz
- a Centre for Advanced Drug Research , COMSATS Institute of Information Technology , Abbottabad , Pakistan
| | - Joanna Lecka
- e Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine , Université Laval , Québec , Canada.,f Centre de Recherche du CHU de Québec , Université Laval , Québec , Canada
| | - Jean Sévigny
- e Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine , Université Laval , Québec , Canada.,f Centre de Recherche du CHU de Québec , Université Laval , Québec , Canada
| | - Chang-Hyun Oh
- g Center for Biomaterials , Korea Institute of Science and Technology , Seoul , Republic of Korea.,h Department of Biomolecular Science , University of Science and Technology , Daejeon , Republic of Korea
| |
Collapse
|
20
|
Stefanachi A, Leonetti F, Pisani L, Catto M, Carotti A. Coumarin: A Natural, Privileged and Versatile Scaffold for Bioactive Compounds. Molecules 2018; 23:E250. [PMID: 29382051 PMCID: PMC6017103 DOI: 10.3390/molecules23020250] [Citation(s) in RCA: 308] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/22/2018] [Accepted: 01/25/2018] [Indexed: 12/22/2022] Open
Abstract
Many naturally occurring substances, traditionally used in popular medicines around the world, contain the coumarin moiety. Coumarin represents a privileged scaffold for medicinal chemists, because of its peculiar physicochemical features, and the versatile and easy synthetic transformation into a large variety of functionalized coumarins. As a consequence, a huge number of coumarin derivatives have been designed, synthesized, and tested to address many pharmacological targets in a selective way, e.g., selective enzyme inhibitors, and more recently, a number of selected targets (multitarget ligands) involved in multifactorial diseases, such as Alzheimer's and Parkinson's diseases. In this review an overview of the most recent synthetic pathways leading to mono- and polyfunctionalized coumarins will be presented, along with the main biological pathways of their biosynthesis and metabolic transformations. The many existing and recent reviews in the field prompted us to make some drastic selections, and therefore, the review is focused on monoamine oxidase, cholinesterase, and aromatase inhibitors, and on multitarget coumarins acting on selected targets of neurodegenerative diseases.
Collapse
Affiliation(s)
- Angela Stefanachi
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona 4, I-70125 Bari, Italy.
| | - Francesco Leonetti
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona 4, I-70125 Bari, Italy.
| | - Leonardo Pisani
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona 4, I-70125 Bari, Italy.
| | - Marco Catto
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona 4, I-70125 Bari, Italy.
| | - Angelo Carotti
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via E. Orabona 4, I-70125 Bari, Italy.
| |
Collapse
|
21
|
Shin S, Im HJ, Kwon YJ, Ye DJ, Baek HS, Kim D, Choi HK, Chun YJ. Human steroid sulfatase induces Wnt/β-catenin signaling and epithelial-mesenchymal transition by upregulating Twist1 and HIF-1α in human prostate and cervical cancer cells. Oncotarget 2017; 8:61604-61617. [PMID: 28977889 PMCID: PMC5617449 DOI: 10.18632/oncotarget.18645] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/22/2017] [Indexed: 12/15/2022] Open
Abstract
Steroid sulfatase (STS) catalyzes the hydrolysis of estrone sulfate and dehydroepiandrosterone sulfate (DHEAS) to their unconjugated biologically active forms. Although STS is considered a therapeutic target for estrogen-dependent diseases, the cellular functions of STS remain unclear. We found that STS induces Wnt/β-catenin s Delete ignaling in PC-3 and HeLa cells. STS increases levels of β-catenin, phospho-β-catenin, and phospho-GSK3β. Enhanced translocation of β-catenin to the nucleus by STS might activate transcription of target genes such as cyclin D1, c-myc, and MMP-7. STS knockdown by siRNA resulted in downregulation of Wnt/β-catenin signaling. β-Catenin/TCF-mediated transcription was also enhanced by STS. STS induced an epithelial-mesenchymal transition (EMT) as it reduced the levels of E-cadherin, whereas levels of mesenchymal markers such as N-cadherin and vimentin were enhanced. We found that STS induced Twist1 expression through HIFα activation as HIF-1α knockdown significantly blocks the ability of STS to induce Twist1 transcription. Furthermore, DHEA, but not DHEAS is capable of inducing Twist1. Treatment with a STS inhibitor prevented STS-mediated Wnt/β-catenin signaling and Twist1 expression. Interestingly, cancer cell migration, invasion, and MMPs expression induced by STS were also inhibited by a STS inhibitor. Taken together, these results suggest that STS induces Wnt/β-catenin signaling and EMT by upregulating Twist1 and HIF-1α. The ability of STS to induce the Wnt/β-catenin signaling and EMT has profound implications on estrogen-mediated carcinogenesis in human cancer cells.
Collapse
Affiliation(s)
- Sangyun Shin
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hee-Jung Im
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yeo-Jung Kwon
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Dong-Jin Ye
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hyoung-Seok Baek
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Donghak Kim
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyung-Kyoon Choi
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Young-Jin Chun
- College of Pharmacy and Center for Metareceptome Research, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
22
|
Quest for steroidomimetics: Amino acids derived steroidal and nonsteroidal architectures. Eur J Med Chem 2017; 133:139-151. [DOI: 10.1016/j.ejmech.2017.03.054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/21/2017] [Accepted: 03/24/2017] [Indexed: 11/18/2022]
|
23
|
Yamaguchi Y, Nishizono N, Kobayashi D, Yoshimura T, Wada K, Oda K. Evaluation of synthesized coumarin derivatives on aromatase inhibitory activity. Bioorg Med Chem Lett 2017; 27:2645-2649. [DOI: 10.1016/j.bmcl.2017.01.062] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/30/2016] [Accepted: 01/19/2017] [Indexed: 01/11/2023]
|
24
|
Ballazhi L, Imeri F, Jashari A, Popovski E, Stojković G, Dimovski AJ, Mikhova B, Mladenovska K. Original research paper. Hydrazinyldiene-chroman-2,4-diones in inducing growth arrest and apoptosis in breast cancer cells: Synergism with doxorubicin and correlation with physicochemical properties. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2017; 67:35-52. [PMID: 28231049 DOI: 10.1515/acph-2017-0006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/21/2016] [Indexed: 11/15/2022]
Abstract
This study evaluates the effects of previously synthesized hydrazinyldiene-chroman-2,4-diones on cell proliferation and apoptosis, cell cycle distribution and migration capacity of MCF-7 breast cancer cells in synergy with doxorubicin. Physicochemical properties of the synthesized compounds were correlated with their structure and activity. Significant cell viability decrease in comparison with the effect of doxorubicin alone and the reference 4-hydroxycoumarin was observed when combination treatment comprising doxorubicin and the title compounds was applied. Synergistic effect with doxorubicin was also observed in down-regulation of phospho-Thr308Akt levels, confirming reduced proliferation and increased apoptosis. Combined treatment increased the percentage of cells arrested at the G2/M stage. Additive inhibition of cell migration was also observed, pointing to the possibility of reducing the risk of metastases. With their solubility profile and log D7.4, all the synthesized compounds follow Lipinski's rule of five for good permeability (absorption) potential.
Collapse
Affiliation(s)
- Lulzime Ballazhi
- Faculty of Pharmacy, Center of Biomolecular Pharmaceutical Analyses University “Ss Cyril and Methodius” , 1000 Skopje , Macedonia (the former Yugoslav Republic of)
| | - Faik Imeri
- Institute of Physiology, University of Zürich , CH-8057, Zürich , Switzerland
| | - Ahmed Jashari
- Faculty of Natural Sciences & Mathematics State University of Tetovo , 1200 Tetovo , Macedonia (the former Yugoslav Republic of)
| | - Emil Popovski
- Institute of Chemistry, Faculty of Natural Sciences and Mathematics, University “Ss. Cyril and Methodius” , PO Box 162 1000 Skopje , Macedonia (the former Yugoslav Republic of)
| | - Goran Stojković
- Institute of Chemistry, Faculty of Natural Sciences and Mathematics, University “Ss. Cyril and Methodius” , PO Box 162 1000 Skopje , Macedonia (the former Yugoslav Republic of)
| | - Aleksandar J. Dimovski
- Faculty of Pharmacy, Center of Biomolecular Pharmaceutical Analyses University “Ss Cyril and Methodius” , 1000 Skopje , Macedonia (the former Yugoslav Republic of)
| | - Bozhana Mikhova
- Bulgarian Academy of Sciences Institute of Organic Chemistry with Centre of Phytochemistry , 1113 Sofia , Bulgaria
| | - Kristina Mladenovska
- Faculty of Pharmacy, Center of Biomolecular Pharmaceutical Analyses University “Ss Cyril and Methodius” , 1000 Skopje , Macedonia (the former Yugoslav Republic of)
| |
Collapse
|
25
|
Jameera Begam A, Jubie S, Nanjan MJ. Estrogen receptor agonists/antagonists in breast cancer therapy: A critical review. Bioorg Chem 2017; 71:257-274. [PMID: 28274582 DOI: 10.1016/j.bioorg.2017.02.011] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 02/15/2017] [Accepted: 02/18/2017] [Indexed: 01/25/2023]
Abstract
Estrogens display intriguing tissue selective action that is of great biomedical importance in the development of optimal therapeutics for the prevention and treatment of breast cancer. There are also strong evidences to show that both endogenous and exogenous estrogens are involved in the pathogenesis of breast cancer. Tamoxifen has been the only drug of choice for more than 30years to treat patients with estrogen related (ER) positive breast tumors. There is a need therefore, for identifying newer, potential and novel candidates for breast cancer. Keeping this in view, the present review focuses on selective estrogen receptor modulators and estrogen antagonists such as sulfatase and aromatase inhibitors involved in breast cancer therapy. A succinct and critical overview of the structure of estrogen receptors, their signaling and involvement in breast carcinogenesis are herein described.
Collapse
Affiliation(s)
- A Jameera Begam
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Udhagamandalam, India; A Constituent College of JSS University, Mysore, India
| | - S Jubie
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Udhagamandalam, India; A Constituent College of JSS University, Mysore, India.
| | - M J Nanjan
- TIFAC CORE HD, JSS University, Mysore, India
| |
Collapse
|
26
|
Mohareb RM, Megally Abdo NY. Synthesis and Cytotoxic Evaluation of Pyran, Dihydropyridine and Thiophene Derivatives of 3-Acetylcoumarin. Chem Pharm Bull (Tokyo) 2016; 63:678-87. [PMID: 26329861 DOI: 10.1248/cpb.c15-00115] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A series of coumarin analogues bearing 4H-pyran rings 2a-d, 11a-d and 1,4-dihydropyridine rings 3a-d, 12a-d at position 3 were synthesized starting from either 3-acetyl coumarin (1) or the coumarin acetohydrazide derivative 4. Condensation of 3-acetylcoumarin (1) with 2-cyanoacetohydrazide afforded 2-cyano-N'-{1-[2-oxo-2H-chromen-3-yl]ethylidene}acetohydrazide (4). Reaction of compound 4 with elemental sulfur and either malononitrile or ethyl cyanoacetate afforded the thiophene derivatives 8 and 9, respectively. The structures of the newly synthesized compounds were confirmed on the basis of their spectral data and elemental analyses. All synthesized compounds were screened for their in vitro anticancer activity against six human cancer cell lines and normal fibroblasts. Several compounds showed potent inhibition with an IC50 value of ˂870 nM. Compound 3d exhibited equivalent cytotoxic effect as the standard CHS 828 against a breast cancer cell line (IC50 value=18 nM). Normal fibroblast cells (WI38) were affected to a much lesser extent (IC50 value >10000 nM).
Collapse
|
27
|
Recent developments of C-4 substituted coumarin derivatives as anticancer agents. Eur J Med Chem 2016; 119:141-68. [PMID: 27155469 DOI: 10.1016/j.ejmech.2016.03.087] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/28/2016] [Accepted: 03/31/2016] [Indexed: 01/01/2023]
Abstract
Cancer is a prominent cause of death in global. Currently, the numbers of drugs that are in clinical practice are having a high prevalence of side effect and multidrug resistance. Researchers have made an attempt to expand a suitable anticancer drug that has no MDR and side effect. Coumarin scaffold became an attractive subject due to their broad spectrum of pharmacological activities. Coumarin derivatives extensively explored for anticancer activities as it possesses minimum side effect along with multi-drug reversal activity. Coumarin derivatives can act by various mechanisms on different tumor cell lines depending on substitution pattern of the core structure of coumarin. Substitution on coumarin nucleus leads to the search for more potent compounds. In this review, we have made an effort to give a synthetic strategy for the preparation of C-4 substituted coumarin derivatives as anticancer agents based on their mechanism of action and also discuss the SAR of the most active compound.
Collapse
|
28
|
Shah R, Singh J, Singh D, Jaggi AS, Singh N. Sulfatase inhibitors for recidivist breast cancer treatment: A chemical review. Eur J Med Chem 2016; 114:170-90. [PMID: 26974384 DOI: 10.1016/j.ejmech.2016.02.054] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 02/17/2016] [Accepted: 02/22/2016] [Indexed: 12/14/2022]
Abstract
Steroid sulfatase (STS) plays a momentous role in the conversion of sulfated steroids, which are biologically inactive, into biologically active un-sulfated steroid hormones, which support the development and growth of a number of hormone-dependent cancers, including breast cancer. Therefore, inhibitors of STS are supposed to be potential drugs for the treatment of breast and other steroid-dependent cancers. The present review concentrates on broad chemical classification of steroid sulfatase inhibitors. The inhibitors reviewed are classified into four main categories: Steroid sulfamate based inhibitors; Steroid non-sulfamate based inhibitors; Non-steroidal sulfamate based inhibitors; Non-steroidal non-sulfamate based inhibitors. A succinct overview of current treatment of cancer, estradiol precursors, STS enzyme and its role in breast cancer is herein described.
Collapse
Affiliation(s)
- Ramanpreet Shah
- Department of Pharmaceutical Sciences and Drug Research, Pharmaceutical Chemistry Research Lab, Punjabi University, Patiala, 147002, India
| | - Jatinder Singh
- Department of Pharmaceutical Sciences and Drug Research, Pharmaceutical Chemistry Research Lab, Punjabi University, Patiala, 147002, India
| | - Dhandeep Singh
- Department of Pharmaceutical Sciences and Drug Research, Pharmaceutical Chemistry Research Lab, Punjabi University, Patiala, 147002, India.
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Pharmaceutical Chemistry Research Lab, Punjabi University, Patiala, 147002, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Pharmaceutical Chemistry Research Lab, Punjabi University, Patiala, 147002, India
| |
Collapse
|
29
|
Rižner TL. The Important Roles of Steroid Sulfatase and Sulfotransferases in Gynecological Diseases. Front Pharmacol 2016; 7:30. [PMID: 26924986 PMCID: PMC4757672 DOI: 10.3389/fphar.2016.00030] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/03/2016] [Indexed: 01/08/2023] Open
Abstract
Gynecological diseases such as endometriosis, adenomyosis and uterine fibroids, and gynecological cancers including endometrial cancer and ovarian cancer, affect a large proportion of women. These diseases are estrogen dependent, and their progression often depends on local estrogen formation. In peripheral tissues, estrogens can be formed from the inactive precursors dehydroepiandrosterone sulfate and estrone sulfate. Sulfatase and sulfotransferases have pivotal roles in these processes, where sulfatase hydrolyzes estrone sulfate to estrone, and dehydroepiandrosterone sulfate to dehydroepiandrosterone, and sulfotransferases catalyze the reverse reactions. Further activation of estrone to the most potent estrogen, estradiol, is catalyzed by 17-ketosteroid reductases, while estradiol can also be formed from dehydroepiandrosterone by the sequential actions of 3β-hydroxysteroid dehydrogenase-Δ4-isomerase, aromatase, and 17-ketosteroid reductase. This review introduces the sulfatase and sulfotransferase enzymes, in terms of their structures and reaction mechanisms, and the regulation and different transcripts of their genes, together with the importance of their currently known single nucleotide polymorphisms. Data on expression of sulfatase and sulfotransferases in gynecological diseases are also reviewed. There are often unchanged mRNA and protein levels in diseased tissue, with higher sulfatase activities in cancerous endometrium, ovarian cancer cell lines, and adenomyosis. This can be indicative of a disturbed balance between the sulfatase and sulfotransferases enzymes, defining the potential for sulfatase as a drug target for treatment of gynecological diseases. Finally, clinical trials with sulfatase inhibitors are discussed, where two inhibitors have already concluded phase II trials, although so far with no convincing clinical outcomes for patients with endometrial cancer and endometriosis.
Collapse
Affiliation(s)
- Tea Lanišnik Rižner
- Faculty of Medicine, Institute of Biochemistry, University of Ljubljana Ljubljana, Slovenia
| |
Collapse
|
30
|
El-Gamal MI, Baek D, Oh CH. A New Series of Cycloalkane-fused Coumarin Sulfonates: Synthesis andIn VitroAntiproliferative Screening. B KOREAN CHEM SOC 2016. [DOI: 10.1002/bkcs.10651] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Mohammed I. El-Gamal
- Department of Medicinal Chemistry; Faculty of Pharmacy, University of Mansoura; Mansoura 35516 Egypt
- Department of Medicinal Chemistry; College of Pharmacy, University of Sharjah; Sharjah 27272 United Arab Emirates
- Sharjah Institute of Medical Research; University of Sharjah; Sharjah 27272 United Arab Emirates
| | - Daejin Baek
- Department of Chemistry; Hanseo University; Seosan 356-706 Republic of Korea
| | - Chang-Hyun Oh
- Center for Biomaterials; Korea Institute of Science and Technology (KIST); Seoul 130-650 Republic of Korea
- Department of Biomolecular Science; University of Science and Technology (UST); Daejeon 305-333 Republic of Korea
| |
Collapse
|
31
|
Sahu PK. Eco-friendly grinding synthesis of a double-layered nanomaterial and the correlation between its basicity, calcination and catalytic activity in the green synthesis of novel fused pyrimidines. RSC Adv 2016. [DOI: 10.1039/c6ra08111h] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The synthesis of hydrotalcite using Al : Mg molar ratios of 1.0 : 3.0 by a grinding method at room temperature is reported.
Collapse
Affiliation(s)
- Pramod K. Sahu
- School of Studies in Chemistry
- Jiwaji University
- Gwalior-474011
- India
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Probing the surface of human carbonic anhydrase for clues towards the design of isoform specific inhibitors. BIOMED RESEARCH INTERNATIONAL 2015; 2015:453543. [PMID: 25811028 PMCID: PMC4355338 DOI: 10.1155/2015/453543] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 09/01/2014] [Indexed: 11/17/2022]
Abstract
The alpha carbonic anhydrases (α-CAs) are a group of structurally related zinc metalloenzymes that catalyze the reversible hydration of CO2 to HCO3−. Humans have 15 different α-CAs with numerous physiological roles and expression patterns. Of these, 12 are catalytically active, and abnormal expression and activities are linked with various diseases, including glaucoma and cancer. Hence there is a need for CA isoform specific inhibitors to avoid off-target CA inhibition, but due to the high amino acid conservation of the active site and surrounding regions between each enzyme, this has proven difficult. However, residues towards the exit of the active site are variable and can be exploited to design isoform selective inhibitors. Here we discuss and characterize this region of “selective drug targetability” and how these observations can be utilized to develop isoform selective CA inhibitors.
Collapse
|
35
|
Chen CY, Zhang XM, Shi JJ, He J. Synthesis of highly functionalized 3,4-cyclohexane-annelated coumarins. RESEARCH ON CHEMICAL INTERMEDIATES 2015. [DOI: 10.1007/s11164-013-1212-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
36
|
Sommerwerk S, Heller L, Csuk R. Synthesis and Cytotoxic Activity of Pentacyclic Triterpenoid Sulfamates. Arch Pharm (Weinheim) 2015; 348:46-54. [DOI: 10.1002/ardp.201400297] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/13/2014] [Accepted: 10/30/2014] [Indexed: 12/18/2022]
Affiliation(s)
- Sven Sommerwerk
- Martin-Luther-Universit; ä; t Halle-Wittenberg; Bereich Organische Chemie; Halle (Saale) Germany
| | - Lucie Heller
- Martin-Luther-Universit; ä; t Halle-Wittenberg; Bereich Organische Chemie; Halle (Saale) Germany
| | - René Csuk
- Martin-Luther-Universit; ä; t Halle-Wittenberg; Bereich Organische Chemie; Halle (Saale) Germany
| |
Collapse
|
37
|
Synthesis, in vitro antiproliferative activity, and in silico studies of fused tricyclic coumarin sulfonate derivatives. Eur J Med Chem 2014; 84:68-76. [DOI: 10.1016/j.ejmech.2014.06.064] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 06/09/2014] [Accepted: 06/27/2014] [Indexed: 01/03/2023]
|
38
|
Basanagouda M, Jambagi VB, Barigidad NN, Laxmeshwar SS, Devaru V, Narayanachar. Synthesis, structure–activity relationship of iodinated-4-aryloxymethyl-coumarins as potential anti-cancer and anti-mycobacterial agents. Eur J Med Chem 2014; 74:225-33. [DOI: 10.1016/j.ejmech.2013.12.061] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 12/25/2013] [Accepted: 12/31/2013] [Indexed: 12/21/2022]
|
39
|
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
|
40
|
Zhang W, Li Z, Zhou M, Wu F, Hou X, Luo H, Liu H, Han X, Yan G, Ding Z, Li R. Synthesis and biological evaluation of 4-(1,2,3-triazol-1-yl)coumarin derivatives as potential antitumor agents. Bioorg Med Chem Lett 2014; 24:799-807. [DOI: 10.1016/j.bmcl.2013.12.095] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 11/21/2013] [Accepted: 12/23/2013] [Indexed: 12/21/2022]
|
41
|
Wlcek K, Hofstetter L, Stieger B. Transport of estradiol-17β-glucuronide, estrone-3-sulfate and taurocholate across the endoplasmic reticulum membrane: evidence for different transport systems. Biochem Pharmacol 2014; 88:106-18. [PMID: 24406246 PMCID: PMC3969151 DOI: 10.1016/j.bcp.2013.12.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 12/23/2013] [Accepted: 12/23/2013] [Indexed: 11/06/2022]
Abstract
Important reactions of drug metabolism, including UGT mediated glucuronidation and steroidsulfatase mediated hydrolysis of sulfates, take place in the endoplasmic reticulum (ER) of hepatocytes. Consequently, UGT generated glucuronides, like estradiol-17β-glucuronide, have to be translocated back into the cytoplasm to reach their site of excretion. Also steroidsulfatase substrates, including estrone-3-sulfate, have to cross the ER membrane to reach their site of hydrolysis. Based on their physicochemical properties such compounds are not favored for passive diffusion and therefore likely necessitate transport system(s) to cross the ER membrane in either direction. The current study aims to investigate the transport of taurocholate, estradiol-17β-glucuronide, and estrone-3-sulfate in smooth (SER) and rough (RER) endoplasmic reticulum membrane vesicles isolated from Wistar and TR− rat liver. Time-dependent and bidirectional transport was demonstrated for taurocholate, showing higher uptake rates in SER than RER vesicles. For estradiol-17β-glucuronide a fast time-dependent efflux with similar efficiencies from SER and RER but no clear protein-mediated uptake was shown, indicating an asymmetric transport system for this substrate. Estrone-3-sulfate uptake was time-dependent and higher in SER than in RER vesicles. Inhibition of steroidsulfatase mediated estrone-3-sulfate hydrolysis decreased estrone-3-sulfate uptake but had no effect on taurocholate or estradiol-17β-glucuronide transport. Based on inhibition studies and transport characteristics, three different transport mechanisms are suggested to be involved in the transport of taurocholate, estrone-3-sulfate and estradiol-17β-glucuronide across the ER membrane.
Collapse
Affiliation(s)
- Katrin Wlcek
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland.
| | - Lia Hofstetter
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland.
| | - Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland.
| |
Collapse
|
42
|
Hussain MK, Ansari MI, Yadav N, Gupta PK, Gupta AK, Saxena R, Fatima I, Manohar M, Kushwaha P, Khedgikar V, Gautam J, Kant R, Maulik PR, Trivedi R, Dwivedi A, Kumar KR, Saxena AK, Hajela K. Design and synthesis of ERα/ERβ selective coumarin and chromene derivatives as potential anti-breast cancer and anti-osteoporotic agents. RSC Adv 2014. [DOI: 10.1039/c3ra45749d] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
43
|
Abstract
The chemical diversity, binding specificity and propensity to interact with biological targets has inspired many researchers to utilize natural products as molecular probes. Almost all reported carbonic anhydrase inhibitors comprise a zinc binding group in their structure of which the primary sulfonamide moiety (-SO2NH2) is the foremost example and to a lesser extent the primary sulfamate (-O-SO2NH2) and sulfamide (-NH-SO2NH2) groups. Natural products that comprise these zinc binding groups in their structure are however rare and relatively few natural products have been explored as a source for novel carbonic anhydrase inhibitors. This chapter will highlight the recent and growing interest in carbonic anhydrase inhibitors sourced from nature, demonstrating that natural product chemical space presents a rich source of potential alternate chemotypes for the discovery of novel drug-like carbonic anhydrase inhibitors.
Collapse
|
44
|
Spillane W, Malaubier JB. Sulfamic Acid and Its N- and O-Substituted Derivatives. Chem Rev 2013; 114:2507-86. [DOI: 10.1021/cr400230c] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- William Spillane
- School
of Chemistry, National University of Ireland, Galway, University Road, Galway, Ireland
| | - Jean-Baptiste Malaubier
- Manufacturing Science
and
Technology, Roche Ireland Limited, Clarecastle, Co. Clare, Ireland
| |
Collapse
|
45
|
Chen C, Hu J, Zhang X, Shi J, He J. Synthesis and Self-Assembled Helical Supramolecular Polymer of Ethyl 7,8-dihydro-3-hydroxy-9-methyl-7-(4′-nitrophenyl)-6H-dibenzo[c]- pyran-6-one-8-carboxylate. HETEROATOM CHEMISTRY 2013. [DOI: 10.1002/hc.21133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Chaoyue Chen
- School of Chemical Engineering; Anhui University of Science and Technology; No.168, Shungeng Middle Road, Huainan 232001 Anhui People's Republic of China
| | - Jinsong Hu
- School of Chemical Engineering; Anhui University of Science and Technology; No.168, Shungeng Middle Road, Huainan 232001 Anhui People's Republic of China
| | - Xiaomei Zhang
- School of Chemical Engineering; Anhui University of Science and Technology; No.168, Shungeng Middle Road, Huainan 232001 Anhui People's Republic of China
| | - Jianjun Shi
- School of Chemical Engineering; Anhui University of Science and Technology; No.168, Shungeng Middle Road, Huainan 232001 Anhui People's Republic of China
| | - Jie He
- School of Chemical Engineering; Anhui University of Science and Technology; No.168, Shungeng Middle Road, Huainan 232001 Anhui People's Republic of China
| |
Collapse
|
46
|
Sulfatase-activated fluorophores for rapid discrimination of mycobacterial species and strains. Proc Natl Acad Sci U S A 2013; 110:12911-6. [PMID: 23878250 DOI: 10.1073/pnas.1222041110] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Most current diagnostic tests for tuberculosis do not reveal the species or strain of pathogen causing pulmonary infection, which can lead to inappropriate treatment regimens and the spread of disease. Here, we report an assay for mycobacterial strain assignment based on genetically conserved mycobacterial sulfatases. We developed a sulfatase-activated probe, 7-hydroxy-9H-(1,3-dichloro-9,9-dimethylacridin-2-one)-sulfate, that detects enzyme activity in native protein gels, allowing the rapid detection of sulfatases in mycobacterial lysates. This assay revealed that mycobacterial strains have distinct sulfatase fingerprints that can be used to judge both the species and lineage. Our results demonstrate the potential of enzyme-activated probes for rapid pathogen discrimination for infectious diseases.
Collapse
|
47
|
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
|
48
|
Kostova I. Studying plant-derived coumarins for their pharmacological and therapeutic properties as potential anticancer drugs. Expert Opin Drug Discov 2013; 2:1605-18. [PMID: 23488904 DOI: 10.1517/17460441.2.12.1605] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Coumarins have attracted intense interest in recent years because of their diverse pharmacological properties. Among these properties, their anticancer effect was most extensively examined. In this review, their broad range of effects on the tumours as shown by various in vitro and in vivo experiments as well as clinical investigations is discussed. Studies have indicated that coumarins elicit inhibitory effects on cell growth of various carcinoma cell lines and may be potential candidates for cancer therapy. These natural compounds have served as valuable leads for further design and synthesis of more active analogues. In view of the relative simplicity of the coumarin compounds and their mechanism of action, the coumarin pharmacophore may serve as an important model on which to develop new patterns in cancer chemotherapy. The aim of this review is to examine in detail the properties of the title compounds as anticancer agents. In view of their comparatively low toxicity, relative cheapness, presence in the diet and occurrence in various herbal remedies, it appears important to evaluate their anticancer potentialities. Moreover their synergistic activity in combination therapy with other well-known anticancer drugs could be the basis for the development of rational approaches to new forms of cancer chemotherapy.
Collapse
Affiliation(s)
- Irena Kostova
- Medical University, Department of Chemistry, Faculty of Pharmacy, 2 Dunav Street, Sofi a 1000, Bulgaria +35 92 92 36 569 ; +35 92 98 79 874 ;
| |
Collapse
|
49
|
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: 3.0] [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
|
50
|
Ouellet É, Maltais R, Ouellet C, Poirier D. Investigation of a tetrahydroisoquinoline scaffold as dual-action steroid sulfatase inhibitors generated by parallel solid-phase synthesis. MEDCHEMCOMM 2013. [DOI: 10.1039/c3md20354a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|