1
|
Kaur J, Bhardwaj A, Wuest F. Fluorine-18 Labelled Radioligands for PET Imaging of Cyclooxygenase-2. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123722. [PMID: 35744851 PMCID: PMC9227202 DOI: 10.3390/molecules27123722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/18/2022]
Abstract
Molecular imaging probes enable the early and accurate detection of disease-specific biomarkers and facilitate personalized treatment of many chronic diseases, including cancer. Among current clinically used functional imaging modalities, positron emission tomography (PET) plays a significant role in cancer detection and in monitoring the response to therapeutic interventions. Several preclinical and clinical studies have demonstrated the crucial involvement of cyclooxygenase-2 (COX-2) isozyme in cancer development and progression, making COX-2 a promising cancer biomarker. A variety of COX-2-targeting PET radioligands has been developed based on anti-inflammatory drugs and selective COX-2 inhibitors. However, many of those suffer from non-specific binding and insufficient metabolic stability. This article highlights examples of COX-2-targeting PET radioligands labelled with the short-lived positron emitter 18F, including radiosynthesis and PET imaging studies published in the last decade (2012–2021).
Collapse
Affiliation(s)
- Jatinder Kaur
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
- Correspondence: (J.K.); (F.W.)
| | - Atul Bhardwaj
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Frank Wuest
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 1Z2, Canada
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 1Z2, Canada
- Correspondence: (J.K.); (F.W.)
| |
Collapse
|
2
|
Kaur J, Bhardwaj A, Wuest F. Development of Fluorescence Imaging Probes for Labeling COX-1 in Live Ovarian Cancer Cells. ACS Med Chem Lett 2021; 12:798-804. [PMID: 34055228 DOI: 10.1021/acsmedchemlett.1c00065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/07/2021] [Indexed: 12/15/2022] Open
Abstract
Recent experimental evidence demonstrated an aberrant overexpression of cyclooxygenase-1 (COX-1) in various cancers, which has stimulated the development of COX-1-selective inhibitors as promising anticancer drugs and cancer imaging agents. Herein we describe the synthesis and validation of 3-(furan-2-yl)-N-aryl 5-amino-pyrazoles as a novel class of COX-1 inhibitors, including molecular docking studies. Among all tested compounds, 4-(5-azido-3-(furan-2-yl)-1H-pyrazol-1-yl)benzoic 17 displayed a favorable COX-1 inhibition and selectivity profile (COX-1 IC50 = 0.1 μM, SI >1000 over COX-2). Compound 17 was selected as a lead structure for developing the novel COX-1-selective fluorescent probe 22. Fluorescent probe 22 was prepared via click chemistry by installing a nitro-benzoxadiazole motif as a fluorophore into the 3-(furan-2-yl)-N-aryl 5-amino-pyrazole scaffold. Fluorescence probe 22 was tested in ovarian cancer cell line OVCAR-3, confirming its usefulness for targeting and visualizing COX-1 in living cells with confocal microscopy.
Collapse
Affiliation(s)
- Jatinder Kaur
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences University of Alberta, 8613 - 114 St., Edmonton, Alberta T6G 2H7, Canada
| | - Atul Bhardwaj
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences University of Alberta, 8613 - 114 St., Edmonton, Alberta T6G 2H7, Canada
| | - Frank Wuest
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton, Alberta T6G 1Z2, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences University of Alberta, 8613 - 114 St., Edmonton, Alberta T6G 2H7, Canada
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Drive, Edmonton, Alberta T6G 2G2, Canada
| |
Collapse
|
3
|
Rouzer CA, Marnett LJ. Structural and Chemical Biology of the Interaction of Cyclooxygenase with Substrates and Non-Steroidal Anti-Inflammatory Drugs. Chem Rev 2020; 120:7592-7641. [PMID: 32609495 PMCID: PMC8253488 DOI: 10.1021/acs.chemrev.0c00215] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cyclooxgenases are key enzymes of lipid signaling. They carry out the first step in the production of prostaglandins, important mediators of inflammation, pain, cardiovascular disease, and cancer, and they are the molecular targets for nonsteroidal anti-inflammatory drugs, which are among the oldest and most chemically diverse set of drugs known. Homodimeric proteins that behave as allosterically modulated, functional heterodimers, the cyclooxygenases exhibit complex kinetic behavior, requiring peroxide-dependent activation and undergoing suicide inactivation. Due to their important physiological and pathophysiological roles and keen interest on the part of the pharmaceutical industry, the cyclooxygenases have been the focus of a vast array of structural studies, leading to the publication of over 80 crystal structures of the enzymes in complex with substrates or inhibitors supported by a wealth of functional data generated by site-directed mutation experiments. In this review, we explore the chemical biology of the cyclooxygenases through the lens of this wealth of structural and functional information. We identify key structural features of the cyclooxygenases, break down their active site into regional binding pockets to facilitate comparisons between structures, and explore similarities and differences in the binding modes of the wide variety of ligands (both substrates and inhibitors) that have been characterized in complex with the enzymes. Throughout, we correlate structure with function whenever possible. Finally, we summarize what can and cannot be learned from the currently available structural data and discuss the critical intriguing questions that remain despite the wealth of information that has been amassed in this field.
Collapse
Affiliation(s)
- Carol A Rouzer
- A. B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Lawrence J Marnett
- A. B. Hancock Jr. Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry, and Pharmacology, Vanderbilt Institute of Chemical Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
4
|
Singh P, Shrestha S, Cortes-Salva MY, Jenko KJ, Zoghbi SS, Morse CL, Innis RB, Pike VW. 3-Substituted 1,5-Diaryl-1 H-1,2,4-triazoles as Prospective PET Radioligands for Imaging Brain COX-1 in Monkey. Part 1: Synthesis and Pharmacology. ACS Chem Neurosci 2018; 9:2610-2619. [PMID: 29678105 DOI: 10.1021/acschemneuro.8b00102] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cyclooxygenase-1 (COX-1) is a key enzyme in the biosynthesis of proinflammatory thromboxanes and prostaglandins and is found in glial and neuronal cells within brain. COX-1 expression is implicated in numerous neuroinflammatory states. We aim to find a direct-acting positron emission tomography (PET) radioligand for imaging COX-1 in human brain as a potential biomarker of neuroinflammation and for serving as a tool in drug development. Seventeen 3-substituted 1,5-diaryl-1 H-1,2,4-triazoles were prepared as prospective COX-1 PET radioligands. From this set, three 1,5-(4-methoxyphenyl)-1 H-1,2,4-triazoles, carrying a 3-methoxy (5), 3-(1,1,1-trifluoroethoxy) (20), or 3-fluoromethoxy substituent (6), were selected for radioligand development, based mainly on their high affinities and selectivities for inhibiting human COX-1, absence of carboxyl group, moderate computed lipophilicities, and scope for radiolabeling with carbon-11 ( t1/2 = 20.4 min) or fluorine-18 ( t1/2 = 109.8 min). Methods were developed for producing [11C]5, [11C]20, and [ d2-18F]6 from hydroxy precursors in a form ready for intravenous injection for prospective evaluation in monkey with PET.
Collapse
Affiliation(s)
- Prachi Singh
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Building 10, Room B3 C346A, 10 Center Drive, Bethesda, Maryland 20892, United States
| | - Stal Shrestha
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Building 10, Room B3 C346A, 10 Center Drive, Bethesda, Maryland 20892, United States
| | - Michelle Y. Cortes-Salva
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Building 10, Room B3 C346A, 10 Center Drive, Bethesda, Maryland 20892, United States
| | - Kimberly J. Jenko
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Building 10, Room B3 C346A, 10 Center Drive, Bethesda, Maryland 20892, United States
| | - Sami S. Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Building 10, Room B3 C346A, 10 Center Drive, Bethesda, Maryland 20892, United States
| | - Cheryl L. Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Building 10, Room B3 C346A, 10 Center Drive, Bethesda, Maryland 20892, United States
| | - Robert B. Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Building 10, Room B3 C346A, 10 Center Drive, Bethesda, Maryland 20892, United States
| | - Victor W. Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Building 10, Room B3 C346A, 10 Center Drive, Bethesda, Maryland 20892, United States
| |
Collapse
|
5
|
Sakata C, Kawasaki T, Iwatsuki Y, Moritani Y, Morita Y, Hara H. Inhibitory effects of ASP6537, a selective cyclooxygenase-1 inhibitor, on thrombosis and neointima formation in rats. Thromb Res 2017; 157:72-78. [PMID: 28700965 DOI: 10.1016/j.thromres.2017.06.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/20/2017] [Accepted: 06/30/2017] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Percutaneous coronary interventions (PCIs), such as balloon angioplasty and stent placement, are effective in the treatment of coronary artery disease. PCI has drawbacks, however, including acute thrombosis after the procedure and restenosis of the vascular lumen due to abnormal neointimal hyperplasia. ASP6537 is a selective COX-1 inhibitor that has been investigated as a possible candidate for clinical development as an antiplatelet agent. In the present study, we evaluated the in vivo antithrombotic effect of ASP6537 and its effect on neointima formation after balloon angioplasty. MATERIAL AND METHODS The antithrombotic effect of ASP6537 was examined using an arteriovenous shunt thrombosis model in rats while the effect of ASP6537 on neointima formation was evaluated in a rat carotid arterial balloon angioplasty model. RESULTS In the thrombosis study, ASP6537 dose-dependently decreased the protein content of the thrombus, while no prolongation of template bleeding time was observed. In the neointima study, ASP6537 reduced neointima formation. CONCLUSIONS ASP6537 may be a promising agent for the prevention of acute thrombosis and restenosis after PCI in place of aspirin.
Collapse
Affiliation(s)
- Chinatsu Sakata
- Project Management, Development, Astellas Pharma Inc., 2-5-1 Nihonbashi Honcho, Chuo-ku, Tokyo 103-8411, Japan.
| | - Tomihisa Kawasaki
- Project Management, Development, Astellas Pharma Inc., 2-5-1 Nihonbashi Honcho, Chuo-ku, Tokyo 103-8411, Japan
| | - Yoshiyuki Iwatsuki
- Pharmacovigilance, Astellas Pharma Inc., 2-5-1 Nihonbashi Honcho, Chuo-ku, Tokyo 103-8411, Japan
| | - Yumiko Moritani
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Yoshiaki Morita
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba-shi, Ibaraki 305-8585, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Gifu Pharmaceutical University, 1-25-4 Daigakunishi, Gifu-shi, Gifu 501-1196, Japan
| |
Collapse
|
6
|
Additive antithrombotic effect of ASP6537, a selective cyclooxygenase (COX)-1 inhibitor, in combination with clopidogrel in guinea pigs. Eur J Pharmacol 2017; 798:72-76. [PMID: 28095326 DOI: 10.1016/j.ejphar.2017.01.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 01/13/2017] [Accepted: 01/13/2017] [Indexed: 11/21/2022]
Abstract
Clopidogrel (Plavix®, Sanofi-Aventis), the adenosine diphosphate P2Y12 receptor antagonist, is reported to be effective in the prevention of cardiovascular events and is often used in combination with aspirin, particularly in high-risk patients. ASP6537 is a reversible cyclooxygenase (COX)-1 inhibitor that is under investigation as an anti-platelet agent. First, we investigated the reversibility of the antiplatelet effect of ASP6537 and its interaction with ibuprofen to compare the usability of ASP6537 with that of aspirin. We then evaluated the antithrombotic effect of ASP6537 in combination with clopidogrel using a FeCl3-induced thrombosis model in guinea pigs. ASP6537 exerted reversible antiplatelet activity, and no pharmacodynamic interaction with ibuprofen was noted. When administered as monotherapy, ASP6537 exerted a significant antithrombotic effect at ≥3mg/kg, while aspirin inhibited thrombosis at 100mg/kg. ASP6537 exerted significant additive effects in combination with clopidogrel, and the minimum antithrombotic dose was reduced by concomitant administration of clopidogrel. Our study showed that ASP6537 did not interact with ibuprofen and has clear additive effects in combination with clopidogrel. ASP6537 may therefore represent a promising antiplatelet agent for use in clinical settings in combination with clopidogrel.
Collapse
|
7
|
Vitale P, Panella A, Scilimati A, Perrone MG. COX-1 Inhibitors: Beyond Structure Toward Therapy. Med Res Rev 2016; 36:641-71. [DOI: 10.1002/med.21389] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 01/21/2016] [Accepted: 02/15/2016] [Indexed: 01/22/2023]
Affiliation(s)
- Paola Vitale
- Department of Pharmacy - Pharmaceutical Sciences; University of Bari “A. Moro”; 70125 Bari Italy
| | - Andrea Panella
- Department of Pharmacy - Pharmaceutical Sciences; University of Bari “A. Moro”; 70125 Bari Italy
| | - Antonio Scilimati
- Department of Pharmacy - Pharmaceutical Sciences; University of Bari “A. Moro”; 70125 Bari Italy
| | - Maria Grazia Perrone
- Department of Pharmacy - Pharmaceutical Sciences; University of Bari “A. Moro”; 70125 Bari Italy
| |
Collapse
|
8
|
Konkle ME, Blobaum AL, Moth CW, Prusakiewicz JJ, Xu S, Ghebreselasie K, Akingbade D, Jacobs AT, Rouzer CA, Lybrand TP, Marnett LJ. Conservative Secondary Shell Substitution In Cyclooxygenase-2 Reduces Inhibition by Indomethacin Amides and Esters via Altered Enzyme Dynamics. Biochemistry 2015; 55:348-59. [PMID: 26704937 PMCID: PMC4721528 DOI: 10.1021/acs.biochem.5b01222] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The cyclooxygenase enzymes (COX-1 and COX-2) are the therapeutic targets of nonsteroidal anti-inflammatory drugs (NSAIDs). Neutralization of the carboxylic acid moiety of the NSAID indomethacin to an ester or amide functionality confers COX-2 selectivity, but the molecular basis for this selectivity has not been completely revealed through mutagenesis studies and/or X-ray crystallographic attempts. We expressed and assayed a number of divergent secondary shell COX-2 active site mutants and found that a COX-2 to COX-1 change at position 472 (Leu in COX-2, Met in COX-1) reduced the potency of enzyme inhibition by a series of COX-2-selective indomethacin amides and esters. In contrast, the potencies of indomethacin, arylacetic acid, propionic acid, and COX-2-selective diarylheterocycle inhibitors were either unaffected or only mildly affected by this mutation. Molecular dynamics simulations revealed identical equilibrium enzyme structures around residue 472; however, calculations indicated that the L472M mutation impacted local low-frequency dynamical COX constriction site motions by stabilizing the active site entrance and slowing constriction site dynamics. Kinetic analysis of inhibitor binding is consistent with the computational findings.
Collapse
Affiliation(s)
- Mary E Konkle
- Departments of Biochemistry, ‡Chemistry, and §Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Structural Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine , Nashville Tennessee 37232-0146, United States
| | - Anna L Blobaum
- Departments of Biochemistry, ‡Chemistry, and §Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Structural Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine , Nashville Tennessee 37232-0146, United States
| | - Christopher W Moth
- Departments of Biochemistry, ‡Chemistry, and §Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Structural Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine , Nashville Tennessee 37232-0146, United States
| | - Jeffery J Prusakiewicz
- Departments of Biochemistry, ‡Chemistry, and §Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Structural Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine , Nashville Tennessee 37232-0146, United States
| | - Shu Xu
- Departments of Biochemistry, ‡Chemistry, and §Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Structural Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine , Nashville Tennessee 37232-0146, United States
| | - Kebreab Ghebreselasie
- Departments of Biochemistry, ‡Chemistry, and §Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Structural Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine , Nashville Tennessee 37232-0146, United States
| | - Dapo Akingbade
- Departments of Biochemistry, ‡Chemistry, and §Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Structural Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine , Nashville Tennessee 37232-0146, United States
| | - Aaron T Jacobs
- Departments of Biochemistry, ‡Chemistry, and §Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Structural Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine , Nashville Tennessee 37232-0146, United States
| | - Carol A Rouzer
- Departments of Biochemistry, ‡Chemistry, and §Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Structural Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine , Nashville Tennessee 37232-0146, United States
| | - Terry P Lybrand
- Departments of Biochemistry, ‡Chemistry, and §Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Structural Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine , Nashville Tennessee 37232-0146, United States
| | - Lawrence J Marnett
- Departments of Biochemistry, ‡Chemistry, and §Pharmacology, Vanderbilt Institute of Chemical Biology, Center for Structural Biology, Center in Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine , Nashville Tennessee 37232-0146, United States
| |
Collapse
|
9
|
Perrone MG, Vitale P, Panella A, Fortuna CG, Scilimati A. General role of the amino and methylsulfamoyl groups in selective cyclooxygenase(COX)-1 inhibition by 1,4-diaryl-1,2,3-triazoles and validation of a predictive pharmacometric PLS model. Eur J Med Chem 2015; 94:252-64. [DOI: 10.1016/j.ejmech.2015.02.049] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 10/23/2022]
|
10
|
Uddin MJ, Elleman AV, Ghebreselasie K, Daniel CK, Crews BC, Nance KD, Huda T, Marnett LJ. Design of Fluorine-Containing 3,4-Diarylfuran-2(5H)-ones as Selective COX-1 Inhibitors. ACS Med Chem Lett 2014; 5:1254-8. [PMID: 25408841 DOI: 10.1021/ml500344j] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 10/12/2014] [Indexed: 01/29/2023] Open
Abstract
We report the design and synthesis of fluorine-containing cyclooxygenase-1 (COX-1)-selective inhibitors to serve as prototypes for the development of a COX-1-targeted imaging agent. Deletion of the SO2CH3 group of rofecoxib switches the compound from a COX-2- to a COX-1-selective inhibitor, providing a 3,4-diarylfuran-2(5H)-one scaffold for structure-activity relationship studies of COX-1 inhibition. A wide range of fluorine-containing 3,4-diarylfuran-2(5H)-ones were designed, synthesized, and tested for their ability to selectively inhibit COX-1 in purified protein and human cancer cell assays. Compounds containing a fluoro-substituent on the C-3 phenyl ring and a methoxy-substituent on the C-4 phenyl ring of the 3,4-diarylfuran-2(5H)-one scaffold were the best COX-1-selective agents of those evaluated, exhibiting IC50s in the submicromolar range. These compounds provide the foundation for development of an agent to facilitate radiologic imaging of ovarian cancer expressing elevated levels of COX-1.
Collapse
Affiliation(s)
- Md. Jashim Uddin
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Anna V. Elleman
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Kebreab Ghebreselasie
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Cristina K. Daniel
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Brenda C. Crews
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Kellie D. Nance
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Tamanna Huda
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Lawrence J. Marnett
- A. B. Hancock, Jr., Memorial
Laboratory for Cancer Research, Department of Biochemistry, Chemistry
and Pharmacology, Vanderbilt Institute of Chemical Biology, Center
for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
11
|
Magnone M, Scarfì S, Sturla L, Guida L, Cuzzocrea S, Di Paola R, Bruzzone S, Salis A, De Flora A, Zocchi E. Fluridone as a new anti-inflammatory drug. Eur J Pharmacol 2013; 720:7-15. [PMID: 24211328 DOI: 10.1016/j.ejphar.2013.10.058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 10/21/2013] [Accepted: 10/30/2013] [Indexed: 12/28/2022]
Abstract
Fluridone is a herbicide extensively utilized in agriculture for its documented safety in animals. Fluridone contains a 4(1H)-pyridone and a trifluoromethyl-benzene moiety, which are also present in molecules with analgesic and anti-inflammatory properties. The established absence of adverse effects of Fluridone on animals prompted us to investigate whether it could represent a new anti-inflammatory compound targeting human cells. In stimulated human monocytes, micromolar Fluridone inhibited cyclooxygenase-2 expression and the release of monocyte chemoattractant protein-1 and prostaglandin-E2, to a similar extent as Acetylsalicylic acid. Fluridone also inhibited the proliferation of aortic smooth muscle cells and reduced proliferation and cytokine release by human activated lymphocytes. The mechanism of Fluridone seems to rely on the dose-dependent inhibition of the nuclear translocation of nuclear factor-κB, a transcription factor playing a pivotal role in inflammation. Fluridone also inhibited the release from stimulated human monocytes of abscisic acid, a plant stress hormone recently discovered also in mammalian cells, where it stimulates pro-inflammatory responses. Interestingly, the mechanism of Fluridone's toxicity in plants relies on the inhibition of the enzyme phytoene desaturase, involved in the biosynthetic pathway of ß-carotene, the precursor of absciscic acid in plants. Finally, administration of Fluridone reduced peritoneal inflammation in Zymosan-treated mice. These results suggest that Fluridone could represent a new prototype of anti-inflammatory drug, also active on abscisic acid pro-inflammatory pathway.
Collapse
Affiliation(s)
- Mirko Magnone
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Ofosu FA. Effective, selective and specific inhibition of COX-1 may overcome the “aspirin paradox”. Thromb Res 2013; 132:1-2. [DOI: 10.1016/j.thromres.2013.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 04/08/2013] [Accepted: 04/14/2013] [Indexed: 11/15/2022]
|
13
|
Roginiel AC, Kohut DL, Kaur S, Saleh AMA, Weber T, Geibel P, Singh H, Geibel JP. Effect of NSAIDs on Na⁺/H⁺ exchanger activity in rat colonic crypts. Am J Physiol Cell Physiol 2013; 305:C512-8. [PMID: 23739181 DOI: 10.1152/ajpcell.00303.2012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs; 1) are widely recommended for several acute and chronic conditions. For example, both indomethacin and aspirin are taken for pain relief. Aspirin is also used for prevention of myocardial infarction, and indomethacin can be administered orally or as a suppository for patients with rheumatoid disease and other chronic inflammatory states. However, use of NSAIDs can cause damage to the mucosal barrier surrounding the gastrointestinal (GI) tract, increasing the risk of ulcer formation. While microencapsulation of NSAIDs has been shown to reduce upper GI injury, sustained release in the lower GI tract and colon may cause epithelial erosion due to increased acidification. The use of suppositories has also been linked to rectal and lower GI bleeding. In this study, we investigated the role of NSAIDs aspirin and indomethacin on Na⁺/H⁺ exchanger (NHE) activity in rat colonic crypts. By comparing average rates of pH recovery between control and NSAID perfusion runs, we were able to determine that both aspirin and indomethacin increase hydrogen extrusion into the colonic lumen. Through treatment with 5-ethylisopropyl amiloride (EIPA), amiloride, and zoniporide dihydrochloride, we further demonstrated that indomethacin specifically enhances proton excretion through regulation of apical NHE-3 and NHE-2 and to a lesser extent on basolateral NHE-1 and NHE-4. Our results suggest that clinical exposure to NSAIDs may affect colonic tissue at the site of selected NHE isoforms, resulting in modulation of transport and barrier function.
Collapse
Affiliation(s)
- Aliya C Roginiel
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Sakata C, Kawasaki T, Kato Y, Abe M, Suzuki KI, Ohmiya M, Funatsu T, Morita Y, Okada M. ASP6537, a novel highly selective cyclooxygenase-1 inhibitor, exerts potent antithrombotic effect without "aspirin dilemma". Thromb Res 2013; 132:56-62. [PMID: 23522855 DOI: 10.1016/j.thromres.2013.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 12/03/2012] [Accepted: 03/01/2013] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Aspirin inhibits both the cyclooxygenase (COX)-1-dependent production of thromboxane A2 (TXA2) in platelets and COX-2-dependent production of anti-aggregatory prostaglandin I2 (PGI2) in vessel walls, resulting in "aspirin dilemma." Our objective is to investigate whether ASP6537 can overcome aspirin dilemma and exert a potent antithrombotic effect without a concurrent ulcerogenic effect. METHODS We evaluated the inhibitory effects of ASP6537 on recombinant human COX-1 (rhCOX-1) and rhCOX-2 activities using a COX-1/2 selectivity test. To determine whether ASP6537 induces aspirin dilemma, we examined the effects of ASP6537 on in vitro TXA2 and PGI2 metabolite production from platelets and isolated aorta of guinea pigs, and on plasma concentrations of TXA2 and PGI2 metabolites in aged rats. Finally, we evaluated the antithrombotic effects and ulcerogenic activity of ASP6537 using an electrically induced carotid arterial thrombosis model and a gastric ulcer model in guinea pigs. RESULTS The IC50 ratios of rhCOX-2 to rhCOX-1 for ASP6537 and aspirin were >142,000 and 1.63 fold, respectively. ASP6537 inhibited TXA2 production more selectively than aspirin in in vitro and in vivo TXA2/PGI2 production studies. ASP6537 exerted a significant antithrombotic effect at ≥3 mg/kg, while aspirin tended to inhibit thrombosis at 300 mg/kg but it was not statistically significant. Further, ASP6537 did not induce ulcer formation at 100 mg/kg, whereas aspirin exhibited an ulcerogenic effect at doses of ≥100 mg/kg. CONCLUSIONS ASP6537 functions as a highly selective COX-1 inhibitor with a superior ability to aspirin for normalizing TXA2/PGI2 balance, and exerts antithrombotic effect without ulcerogenic effect.
Collapse
Affiliation(s)
- Chinatsu Sakata
- Pharmacology Research Labs., Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Perrone MG, Vitale P, Malerba P, Altomare A, Rizzi R, Lavecchia A, Di Giovanni C, Novellino E, Scilimati A. Diarylheterocycle Core Ring Features Effect in Selective COX-1 Inhibition. ChemMedChem 2012; 7:629-41. [DOI: 10.1002/cmdc.201100530] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 12/07/2011] [Indexed: 11/06/2022]
|