1
|
Mondal S, Mondal S, Saha A. Thiuram Disulfide Mediated Cu-Catalyzed Amidation of Terminal Alkynes: An Efficient Synthesis of Alkynyl Amides. J Org Chem 2024; 89:2182-2189. [PMID: 38326283 DOI: 10.1021/acs.joc.3c01961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Terminal alkynes undergo a CO-free aminocarbonylation reaction mediated by thiuram disulfides. Thiuram disulfide acts as the source of the carbamoyl group in the amidation of terminal alkynes in the presence of copper-based reagent and catalyst. A series of alkynyl amides has been prepared with several structural variations following the current one-pot two-step protocol. The reaction proceeds through a mixed disulfide intermediate, which has been isolated and characterized by single-crystal XRD analysis.
Collapse
Affiliation(s)
- Sourav Mondal
- Department of Chemistry, Jadavpur University, Kolkata 700032, India
| | - Soumya Mondal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Amit Saha
- Department of Chemistry, Jadavpur University, Kolkata 700032, India
| |
Collapse
|
2
|
Chandrabalan A, Ramachandran R. Molecular mechanisms regulating Proteinase‐Activated Receptors (PARs). FEBS J 2021; 288:2697-2726. [DOI: 10.1111/febs.15829] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Arundhasa Chandrabalan
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry University of Western Ontario London Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry University of Western Ontario London Canada
| |
Collapse
|
3
|
Liu S, Li S, Yuan D, Wang E, Xie R, Zhang W, Kong Y, Zhu X. Protease activated receptor 4 (PAR4) antagonists: Research progress on small molecules in the field of antiplatelet agents. Eur J Med Chem 2020; 209:112893. [PMID: 33049608 DOI: 10.1016/j.ejmech.2020.112893] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 12/27/2022]
Abstract
Protease activated receptor 4 (PAR4) is a key target in antiplatelet medication to reduce the risk of heart attack and thrombotic complications in stroke. PAR4 antagonists can prevent harmful and stable thrombus growth while retaining initial thrombus formation by acting on the late diffusion stage of platelet activation, which may provide a safer alternative than other antiplatelet agents. Currently, research on PAR4 antagonists is of increasing interest in the field of antiplatelet agents. This article provides an overview of the discovery and development of small-molecule antagonists of PAR4 as novel antiplatelet agents, including structure-activity relationship (SAR) analysis, progress of structure and bioassay optimization, and the latest structural and/or clinical information of representative small-molecule antagonists of PAR4.
Collapse
Affiliation(s)
- Shangde Liu
- Institute of Medicinal & Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Shanshan Li
- Institute of Medicinal & Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Duo Yuan
- Institute of Medicinal & Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Enmao Wang
- Institute of Medicinal & Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Roujie Xie
- Institute of Medicinal & Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Weiqi Zhang
- Institute of Medicinal & Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yi Kong
- School of Life & Technology, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Xiong Zhu
- Institute of Medicinal & Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China.
| |
Collapse
|
4
|
Poulie CBM, Liu N, Jensen AA, Bunch L. Design, Synthesis, and Pharmacological Characterization of Heterobivalent Ligands for the Putative 5-HT 2A/mGlu 2 Receptor Complex. J Med Chem 2020; 63:9928-9949. [PMID: 32815361 DOI: 10.1021/acs.jmedchem.0c01058] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We report the synthesis of the first series of heterobivalent ligands targeting the putative heteromeric 5-HT2A/mGlu2 receptor complex, based on the 5-HT2A antagonist MDL-100,907 and the mGlu2 ago-PAM JNJ-42491293. The functional properties of monovalent and heterobivalent ligands were characterized in 5-HT2A-, mGlu2/Gqo5-, 5-HT2A/mGlu2-, and 5-HT2A/mGlu2/Gqo5-expressing HEK293 cells using a Ca2+ imaging assay and a [3H]ketanserin binding assay. Pronounced functional crosstalk was observed between the two receptors in 5-HT2A/mGlu2 and 5-HT2A/mGlu2/Gqo5 cells. While the synthesized monovalent ligands retained the 5-HT2A antagonist and mGlu2 ago-PAM functionalities, the seven bivalent ligands inhibited 5-HT-induced responses in 5-HT2A/mGlu2 cells and both 5-HT- and Glu-induced responses in 5-HT2A/mGlu2/Gqo5 cells. However, no definitive correlation between the functional potency and spacer length of the ligands was observed, an observation substantiated by the binding affinities exhibited by the compounds in 5-HT2A, 5-HT2A/mGlu2, and 5-HT2A/mGlu2/Gqo5 cells. In conclusion, while functional crosstalk between 5-HT2A and mGlu2 was demonstrated, it remains unclear how these heterobivalent ligands interact with the putative receptor complex.
Collapse
Affiliation(s)
- Christian B M Poulie
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen OE, Denmark
| | - Na Liu
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen OE, Denmark
| | - Anders A Jensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen OE, Denmark
| | - Lennart Bunch
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen OE, Denmark
| |
Collapse
|
5
|
Kapoor A, Ghosh AK, Forman M, Hu X, Ye W, Southall N, Marugan J, Keyes RF, Smith BC, Meyers DJ, Ferrer M, Arav-Boger R. Validation and Characterization of Five Distinct Novel Inhibitors of Human Cytomegalovirus. J Med Chem 2020; 63:3896-3907. [PMID: 32191456 PMCID: PMC7386824 DOI: 10.1021/acs.jmedchem.9b01501] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The critical consequences of human cytomegalovirus (HCMV) infection in the transplant population and in congenitally infected infants, the limited treatment options for HCMV, and the rise of resistant mutants toward existing therapies has fueled the search for new anti-HCMV agents. A pp28-luciferase recombinant HCMV was used as a reporter system for high-throughput screening of HCMV inhibitors. Approximately 400 000 compounds from existing libraries were screened. Subsequent validation assays using resynthesized compounds, several virus strains, and detailed virology assays resulted in the identification of five structurally unique and selective HCMV inhibitors, active at sub to low micromolar concentrations. Further characterization revealed that each compound inhibited a specific stage of HCMV replication. One compound was also active against herpes simplex virus (HSV1 and HSV2), and another compound was active against Epstein-Barr virus (EBV). Drug combination studies revealed that all five compounds were additive with ganciclovir or letermovir. Future studies will focus on optimization of these new anti-HCMV compounds along with mechanistic studies.
Collapse
Affiliation(s)
- Arun Kapoor
- Department of Pediatrics, Division of Infectious Disease, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ayan K. Ghosh
- Department of Pediatrics, Division of Infectious Disease, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael Forman
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | - Xin Hu
- National Center for Advancing Translational Sciences (NCATS), National Institute of Health, Bethesda, MD 20850, USA
| | - Wenjuan Ye
- National Center for Advancing Translational Sciences (NCATS), National Institute of Health, Bethesda, MD 20850, USA
| | - Noel Southall
- National Center for Advancing Translational Sciences (NCATS), National Institute of Health, Bethesda, MD 20850, USA
| | - Juan Marugan
- National Center for Advancing Translational Sciences (NCATS), National Institute of Health, Bethesda, MD 20850, USA
| | - Robert F. Keyes
- Department of Biochemistry, Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Brian C. Smith
- Department of Biochemistry, Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - David J. Meyers
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Marc Ferrer
- National Center for Advancing Translational Sciences (NCATS), National Institute of Health, Bethesda, MD 20850, USA
| | - Ravit Arav-Boger
- Department of Pediatrics, Division of Infectious Disease, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
6
|
Fan M, Han M, Xia Y, Zhang Y, Chu Y, Bai G, Li W, Li J, Zhao L, He Y, Ma X, Duan Z. Design and synthesis of potent PAR-1 antagonists based on vorapaxar. Bioorg Med Chem Lett 2020; 30:127046. [PMID: 32122739 DOI: 10.1016/j.bmcl.2020.127046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/11/2020] [Accepted: 02/15/2020] [Indexed: 01/23/2023]
Abstract
A series of novel vorapaxar analogues with different amino substitutes at the C-7, C-9a and aromatic substitutes at the C-4 position were designed, synthesized, and evaluated for their inhibitory activity to PAR-1. Several compounds showed good potency in antagonist activity based on the intracellular calcium mobilization assay and excellent pharmacokinetics profile in rats. Among these analogues, 3d exhibited excellent PAR-1 inhibitory activity (IC50 = 0.18 μM) and the lower ability to cross the blood-brain barrier compared with vorapaxar (IC50 = 0.25 μM). Compound 3d has the potential to be developed as a new generation of PAR-1 antagonists with a better therapeutic window.
Collapse
Affiliation(s)
- Mengna Fan
- Hebei University of Technology, Tianjin 300100, China; Tianjin Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin 300410, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Holding Group Co., Ltd., Tianjin 300410, China
| | - Min Han
- Tianjin Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin 300410, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Holding Group Co., Ltd., Tianjin 300410, China
| | - Yan Xia
- Tianjin Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin 300410, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Holding Group Co., Ltd., Tianjin 300410, China
| | - Yingbin Zhang
- Hebei University of Technology, Tianjin 300100, China; Tianjin Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin 300410, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Holding Group Co., Ltd., Tianjin 300410, China
| | - Yang Chu
- Tianjin Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin 300410, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Holding Group Co., Ltd., Tianjin 300410, China
| | - Guirong Bai
- Tianjin Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin 300410, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Holding Group Co., Ltd., Tianjin 300410, China
| | - Wei Li
- Tianjin Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin 300410, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Holding Group Co., Ltd., Tianjin 300410, China
| | - Ju Li
- Tianjin Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin 300410, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Holding Group Co., Ltd., Tianjin 300410, China
| | - Lihui Zhao
- Tianjin Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin 300410, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Holding Group Co., Ltd., Tianjin 300410, China
| | - Yi He
- Tianjin Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin 300410, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Holding Group Co., Ltd., Tianjin 300410, China
| | - Xiaohui Ma
- Tianjin Tasly Academy, Tasly Holding Group Co., Ltd., Tianjin 300410, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Holding Group Co., Ltd., Tianjin 300410, China
| | - Zhongyu Duan
- Hebei University of Technology, Tianjin 300100, China.
| |
Collapse
|
7
|
Abstract
As basic research into GPCR signaling and its association with disease has come into fruition, greater clarity has emerged with regards to how these receptors may be amenable to therapeutic intervention. As a diverse group of receptor proteins, which regulate a variety of intracellular signaling pathways, research in this area has been slow to yield tangible therapeutic agents for the treatment of a number of diseases including cancer. However, recently such research has gained momentum based on a series of studies that have sought to define GPCR proteins dynamics through the elucidation of their crystal structures. In this chapter, we define the approaches that have been adopted in developing better therapeutics directed against the specific parts of the receptor proteins, such as the extracellular and the intracellular domains, including the ligands and auxiliary proteins that bind them. Finally, we also briefly outline how GPCR-derived signaling transduction pathways hold great potential as additional targets.
Collapse
Affiliation(s)
- Surinder M Soond
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation.
| |
Collapse
|
8
|
Chiaramonte N, Bua S, Angeli A, Ferraroni M, Picchioni I, Bartolucci G, Braconi L, Dei S, Teodori E, Supuran CT, Romanelli MN. Sulfonamides incorporating piperazine bioisosteres as potent human carbonic anhydrase I, II, IV and IX inhibitors. Bioorg Chem 2019; 91:103130. [PMID: 31374520 DOI: 10.1016/j.bioorg.2019.103130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 12/27/2022]
Abstract
Starting from the molecular simplification of (R) 4-(3,4-dibenzylpiperazine-1-carbonyl)benzenesulfonamide 9a, a compound endowed with selectivity for human Carbonic Anhydrase (hCA) IV, a series of piperazines and 4-aminopiperidines carrying a 4-sulfamoylbenzamide moiety as Zn-binding group have been designed and tested on human isoforms hCA I, II, IV and IX, using a stopped flow CO2 hydrase assay. The aim of the work was to derive structure-activity relationships useful for designing isoform selective compounds. These structural modifications changed the selectivity profile of the analogues from hCA IV to hCA I and II, and improved potency. Several of the new compounds showed subnanomolar activity on hCA II. X-ray crystallography of ligand-hCAII complexes was used to compare the binding modes of the new piperazines and the previously synthesized 2-benzyl-piperazine analogues, explaining the inhibition profiles.
Collapse
Affiliation(s)
- Niccolò Chiaramonte
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Silvia Bua
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Andrea Angeli
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Marta Ferraroni
- University of Florence, Department of Chemistry, via della Lastruccia, 50019 Sesto Fiorentino, Italy
| | - Ilaria Picchioni
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Gianluca Bartolucci
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Laura Braconi
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Silvia Dei
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Elisabetta Teodori
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Claudiu T Supuran
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy.
| | - Maria Novella Romanelli
- University of Florence, Department of Neuroscience, Psychology, Drug Research and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy.
| |
Collapse
|
9
|
Heuberger DM, Schuepbach RA. Protease-activated receptors (PARs): mechanisms of action and potential therapeutic modulators in PAR-driven inflammatory diseases. Thromb J 2019; 17:4. [PMID: 30976204 PMCID: PMC6440139 DOI: 10.1186/s12959-019-0194-8] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/08/2019] [Indexed: 12/29/2022] Open
Abstract
Inflammatory diseases have become increasingly prevalent with industrialization. To address this, numerous anti-inflammatory agents and molecular targets have been considered in clinical trials. Among molecular targets, protease-activated receptors (PARs) are abundantly recognized for their roles in the development of chronic inflammatory diseases. In particular, several inflammatory effects are directly mediated by the sensing of proteolytic activity by PARs. PARs belong to the seven transmembrane domain G protein-coupled receptor family, but are unique in their lack of physiologically soluble ligands. In contrast with classical receptors, PARs are activated by N-terminal proteolytic cleavage. Upon removal of specific N-terminal peptides, the resulting N-termini serve as tethered activation ligands that interact with the extracellular loop 2 domain and initiate receptor signaling. In the classical pathway, activated receptors mediate signaling by recruiting G proteins. However, activation of PARs alternatively lead to the transactivation of and signaling through receptors such as co-localized PARs, ion channels, and toll-like receptors. In this review we consider PARs and their modulators as potential therapeutic agents, and summarize the current understanding of PAR functions from clinical and in vitro studies of PAR-related inflammation.
Collapse
Affiliation(s)
- Dorothea M Heuberger
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Surgical Research Division, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Reto A Schuepbach
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
He Y, Wang Y, Liang X, Huang B, Wang H, Pan YM. Palladium-Catalyzed Three-Component Reaction: A Novel Method for the Synthesis of N-Acyl Propiolamides. Org Lett 2018; 20:7117-7120. [PMID: 30362763 DOI: 10.1021/acs.orglett.8b03068] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Palladium-catalyzed three-component reactions between terminal alkynes, isonitriles, and sodium carboxylates have been developed. This novel and operationally simple methodology provides an alternative for the synthesis of N-acyl propiolamide derivatives under mild conditions using isonitriles as the amine source and sodium carboxylates as the oxygen and acyl source.
Collapse
Affiliation(s)
- Yan He
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources , School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University , Guilin 541004 , People's Republic of China
| | - Yingchun Wang
- College of Chemistry and Chemical Engineering , Jishou University , Jishou 416000 , People's Republic of China
| | - Xinping Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources , School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University , Guilin 541004 , People's Republic of China
| | - Bin Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources , School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University , Guilin 541004 , People's Republic of China
| | - Hengshan Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources , School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University , Guilin 541004 , People's Republic of China
| | - Ying-Ming Pan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources , School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University , Guilin 541004 , People's Republic of China
| |
Collapse
|
11
|
Flaumenhaft R, De Ceunynck K. Targeting PAR1: Now What? Trends Pharmacol Sci 2017; 38:701-716. [PMID: 28558960 PMCID: PMC5580498 DOI: 10.1016/j.tips.2017.05.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 05/01/2017] [Accepted: 05/04/2017] [Indexed: 12/30/2022]
Abstract
Protease-activated receptors (PARs) are a ubiquitously expressed class of G-protein-coupled receptors (GPCRs) that enable cells to respond to proteases in the extracellular environment in a nuanced and dynamic manner. PAR1 is the archetypal family member and has been the object of large-scale drug development programs since the 1990s. Vorapaxar and drotrecogin-alfa are approved PAR1-targeted therapeutics, but safety concerns have limited the clinical use of vorapaxar and questions regarding the efficacy of drotrecogin-alfa led to its withdrawal from the market. New understanding of mechanisms of PAR1 function, discovery of improved strategies for modifying PAR1 function, and identification of novel indications for PAR1 modulators have provided new opportunities for therapies targeting PAR1. In this review, we critically evaluate prospects for the next generation of PAR1-targeted therapeutics.
Collapse
Affiliation(s)
- Robert Flaumenhaft
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Karen De Ceunynck
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
12
|
Abstract
Antiplatelet therapy displays a critical role in the treatment and prevention of antithrombotic disorders. Many new antiplatelet agents have been developed following the emergence of various clinical limitations of classical antiplatelet drugs. This review covers mainly the recent advances in the development of P2Y12 antagonists and GPIIb/IIIa antagonists. Meanwhile, it summarizes promising approaches to new platelet surface receptors such as prostanoid EP3 receptor, thromboxane A2 prostanoid receptor, protease-activated receptors, GPIb-IX-V receptor and P-selectin. In addition, PI3Kβ, a critical protein at the inside signaling pathway of platelet activation is also mentioned as an important antiplatelet target. Moreover, the development of respective drug candidates is discussed in detail.
Collapse
|
13
|
Hughes NL, Brown CL, Irwin AA, Cao Q, Muldoon MJ. Palladium(II)-Catalysed Aminocarbonylation of Terminal Alkynes for the Synthesis of 2-Ynamides: Addressing the Challenges of Solvents and Gas Mixtures. CHEMSUSCHEM 2017; 10:675-680. [PMID: 27906507 PMCID: PMC5347853 DOI: 10.1002/cssc.201601601] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Indexed: 06/06/2023]
Abstract
2-Ynamides can be synthesised through PdII catalysed oxidative carbonylation, utilising low catalyst loadings. A variety of alkynes and amines can be used to afford 2-ynamides in high yields, whilst overcoming the drawbacks associated with previous oxidative methods, which rely on dangerous solvents and gas mixtures. The use of [NBu4 ]I allows the utilisation of the industrially recommended solvent ethyl acetate. O2 can be used as the terminal oxidant, and the catalyst can operate under safer conditions with low O2 concentrations.
Collapse
Affiliation(s)
- N. Louise Hughes
- School of Chemistry and Chemical EngineeringQueen's University of BelfastStranmillis RoadBelfast, David Keir BuildingBT9 5AGNorthern Ireland
| | - Clare L. Brown
- School of Chemistry and Chemical EngineeringQueen's University of BelfastStranmillis RoadBelfast, David Keir BuildingBT9 5AGNorthern Ireland
| | - Andrew A. Irwin
- School of Chemistry and Chemical EngineeringQueen's University of BelfastStranmillis RoadBelfast, David Keir BuildingBT9 5AGNorthern Ireland
| | - Qun Cao
- School of Chemistry and Chemical EngineeringQueen's University of BelfastStranmillis RoadBelfast, David Keir BuildingBT9 5AGNorthern Ireland
| | - Mark J. Muldoon
- School of Chemistry and Chemical EngineeringQueen's University of BelfastStranmillis RoadBelfast, David Keir BuildingBT9 5AGNorthern Ireland
| |
Collapse
|
14
|
Wang Y, Liu J, Zhu T, Zhang L, He X, Zhang JZ. Predicted PAR1 inhibitors from multiple computational methods. Chem Phys Lett 2016. [DOI: 10.1016/j.cplett.2016.07.059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
15
|
Monjotin N, Gillespie J, Farrié M, Le Grand B, Junquero D, Vergnolle N. F16357, a novel protease-activated receptor 1 antagonist, improves urodynamic parameters in a rat model of interstitial cystitis. Br J Pharmacol 2016; 173:2224-36. [PMID: 27111354 PMCID: PMC4919585 DOI: 10.1111/bph.13501] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/17/2016] [Accepted: 04/10/2016] [Indexed: 01/15/2023] Open
Abstract
Background and Purpose The aims of the present study were to characterize the role of PAR1 in rat bladder under inflammatory conditions and determine whether a selective PAR1 antagonist, F16357, can prevent the pathophysiological symptoms of cyclophosphamide‐induced interstitial cystitis (IC). Experimental Approach Immunohistochemistry, contractile activity in isolated bladder and urodynamics were determined before and after cyclophosphamide treatment. F16357 was administered intravesically during the acute phase of inflammation, and effects on PAR1 and PAR1‐related bladder contraction evaluated 24 h after cyclophosphamide injection. Urodynamics and associated voided volumes were recorded 7 and 24 h after cyclophosphamide. Key Results In control conditions, PAR1 was present only in some umbrella cells. Cyclophosphamide disrupted the urothelium and expression of PAR1 by all remaining urothelial cells. After F16357 treatment, urothelial damage was absent and PAR1 immunoreactivity similar to control tissues. Thrombin and TFLLR‐NH2 induced bladder contractions. These were increased in inflammatory conditions and antagonized by F16357 in a concentration‐dependent manner. In telemetric experiments, furosemide increased urine production and voiding frequency for 60 min, 7 h after cyclophosphamide injection. Intravesical administration of F16357 blocked these changes with a return to a physiological profile; 24 h after cyclophosphamide, the volume of micturition was still lower with no increase in number of micturitions. F16357 30 μM reduced the number of micturitions and improved bladder capacity, but did not affect diuresis. Under similar experimental conditions, lidocaine 2% induced comparable effects. Conclusions and Implications PAR1 is expressed in rat bladder, overactivated in inflammatory conditions and involved in bladder function and sensation. F16357 could represent an interesting candidate for IC treatment.
Collapse
Affiliation(s)
- N Monjotin
- Institut de Recherche Pierre Fabre, Castres, France
| | - J Gillespie
- Newcastle University, Newcastle upon Tyne, England
| | - M Farrié
- Institut de Recherche Pierre Fabre, Castres, France
| | - B Le Grand
- Institut de Recherche Pierre Fabre, Castres, France
| | - D Junquero
- Institut de Recherche Pierre Fabre, Castres, France
| | - N Vergnolle
- IRSD, Université de Toulouse, INSERM, INRA, INP-ENVT, Université de Toulouse-3 Paul Sabatier, Toulouse, France
| |
Collapse
|
16
|
Obreque-Balboa JE, Sun Q, Bernhardt G, König B, Buschauer A. Flavonoid derivatives as selective ABCC1 modulators: Synthesis and functional characterization. Eur J Med Chem 2015; 109:124-33. [PMID: 26774038 DOI: 10.1016/j.ejmech.2015.12.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 11/23/2015] [Accepted: 12/09/2015] [Indexed: 11/28/2022]
Abstract
A series of chromones, bearing substituted amino groups or N-substituted carboxamide moieties in position 2, was synthesized and characterized in cellular assays for modulation of the ABC transporters ABCC1 (MDCKII-MRP1 cells), ABCB1 (Kb-V1 cells) and ABCG2 (MCF-7/Topo cells). The most potent ABCC1 modulators identified among these flavonoid-type compounds were comparable to the reference compound reversan regarding potency, but superior in terms of selectivity concerning ABCB1 and ABCG2 (2-[4-(Benzo[c][1,2,5]oxadiazol-5-ylmethyl)piperazin-1-yl]-5,7-dimethoxy-4H-chromen-4-one (51): ABCC1, IC50 11.3 μM; inactive at ABCB1 and ABCG2). Compound 51 was as effective as reversan in reverting ABCC1-mediated resistance to cytostatics in MDCKII-MRP1 cells and proved to be stable in mouse plasma and cell culture medium. Modulators, such as compound 51, are of potential value as pharmacological tools for the investigation of the (patho)physiological role of ABCC1.
Collapse
Affiliation(s)
| | - Qiu Sun
- Institute of Organic Chemistry, University of Regensburg, D-93040 Regensburg, Germany
| | - Günther Bernhardt
- Institute of Pharmacy, University of Regensburg, D-93040 Regensburg, Germany
| | - Burkhard König
- Institute of Organic Chemistry, University of Regensburg, D-93040 Regensburg, Germany.
| | - Armin Buschauer
- Institute of Pharmacy, University of Regensburg, D-93040 Regensburg, Germany.
| |
Collapse
|
17
|
Premachandra IDUA, Scott KA, Shen C, Wang F, Lane S, Liu H, Van Vranken DL. Potent Synergy between Spirocyclic Pyrrolidinoindolinones and Fluconazole against Candida albicans. ChemMedChem 2015; 10:1672-86. [PMID: 26263912 PMCID: PMC4682886 DOI: 10.1002/cmdc.201500271] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Indexed: 11/12/2022]
Abstract
A spiroindolinone, (1S,3R,3aR,6aS)-1-benzyl-6'-chloro-5-(4-fluorophenyl)-7'-methylspiro[1,2,3a,6a-tetrahydropyrrolo[3,4-c]pyrrole-3,3'-1H-indole]-2',4,6-trione, was previously reported to enhance the antifungal effect of fluconazole against Candida albicans. A diastereomer of this compound was synthesized, along with various analogues. Many of the compounds were shown to enhance the antifungal effect of fluconazole against C. albicans, some with exquisite potency. One spirocyclic piperazine derivative, which we have named synazo-1, was found to enhance the effect of fluconazole with an EC50 value of 300 pM against a susceptible strain of C. albicans and going as low as 2 nM against some resistant strains. Synazo-1 exhibits true synergy with fluconazole, with an FIC index below 0.5 in the strains tested. Synazo-1 exhibited low toxicity in mammalian cells relative to the concentrations required for antifungal synergy.
Collapse
Affiliation(s)
| | - Kevin A Scott
- Department of Chemistry, University of California, Irvine, 1102 Natural Sciences 2, Irvine, CA 92697-2025 (USA)
| | - Chengtian Shen
- Department of Chemistry, University of California, Irvine, 1102 Natural Sciences 2, Irvine, CA 92697-2025 (USA)
| | - Fuqiang Wang
- Department of Biological Chemistry, University of California, Irvine, 825 Health Sciences Road, Medical Sciences I, Irvine, CA 92697-1700 (USA)
| | - Shelley Lane
- Department of Biological Chemistry, University of California, Irvine, 825 Health Sciences Road, Medical Sciences I, Irvine, CA 92697-1700 (USA)
| | - Haoping Liu
- Department of Biological Chemistry, University of California, Irvine, 825 Health Sciences Road, Medical Sciences I, Irvine, CA 92697-1700 (USA)
| | - David L Van Vranken
- Department of Chemistry, University of California, Irvine, 1102 Natural Sciences 2, Irvine, CA 92697-2025 (USA).
| |
Collapse
|
18
|
Romberger DJ, Heires AJ, Nordgren TM, Souder CP, West W, Liu XD, Poole JA, Toews ML, Wyatt TA. Proteases in agricultural dust induce lung inflammation through PAR-1 and PAR-2 activation. Am J Physiol Lung Cell Mol Physiol 2015; 309:L388-99. [PMID: 26092994 DOI: 10.1152/ajplung.00025.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 06/09/2015] [Indexed: 01/31/2023] Open
Abstract
Workers exposed to aerosolized dust present in concentrated animal feeding operations (CAFOs) are susceptible to inflammatory lung diseases, such as chronic obstructive pulmonary disease. Extracts of dust collected from hog CAFOs [hog dust extract (HDE)] are potent stimulators of lung inflammatory responses in several model systems. The observation that HDE contains active proteases prompted the present study, which evaluated the role of CAFO dust proteases in lung inflammatory processes and tested whether protease-activated receptors (PARs) are involved in the signaling pathway for these events. We hypothesized that the damaging proinflammatory effect of HDE is due, in part, to the proteolytic activation of PARs, and inhibiting the proteases in HDE or disrupting PAR activation would attenuate HDE-mediated inflammatory indexes in bronchial epithelial cells (BECs), in mouse lung slices in vitro, and in a murine in vivo exposure model. Human BECs and mouse lung slice cultures stimulated with 5% HDE released significantly more of each of the cytokines measured (IL-6, IL-8, TNF-α, keratinocyte-derived chemokine/CXC chemokine ligand 1, and macrophage inflammatory protein-2/CXC chemokine ligand 2) than controls, and these effects were markedly diminished by protease inhibition. Inhibition of PARs also blunted the HDE-induced cytokine release from BECs. In addition, protease depletion inhibited HDE-induced BEC intracellular PKCα and PKCε activation. C57BL/6J mice administered 12.5% HDE intranasally, either once or daily for 3 wk, exhibited increased total cellular and neutrophil influx, bronchial alveolar fluid inflammatory cytokines, lung histopathology, and inflammatory scores compared with mice receiving protease-depleted HDE. These data suggest that proteases in dust from CAFOs are important mediators of lung inflammation, and these proteases and their receptors may provide novel targets for therapeutic intervention in CAFO dust-induced airways disease.
Collapse
Affiliation(s)
- Debra J Romberger
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska; Veterans Affairs Nebraska-Western Iowa Healthcare System, Omaha, Nebraska;
| | - Art J Heires
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska
| | - Tara M Nordgren
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska
| | - Chelsea P Souder
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska
| | - William West
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Xiang-de Liu
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jill A Poole
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska
| | - Myron L Toews
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - Todd A Wyatt
- Pulmonary, Critical Care, Sleep and Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska; Veterans Affairs Nebraska-Western Iowa Healthcare System, Omaha, Nebraska; Department of Environmental, Agricultural, and Occupational Health, University of Nebraska Medical Center, Omaha, Nebraska; and
| |
Collapse
|
19
|
Sadeghzadeh M, Sheibani S, Ghandi M, Daha FJ, Amanlou M, Arjmand M, Hasani Bozcheloie A. Synthesis and receptor binding studies of novel 4,4-disubstituted arylalkyl/arylalkylsulfonyl piperazine and piperidine-based derivatives as a new class of σ1 ligands. Eur J Med Chem 2013; 64:488-97. [DOI: 10.1016/j.ejmech.2013.04.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 04/02/2013] [Accepted: 04/03/2013] [Indexed: 11/30/2022]
|
20
|
Yan J, Manaenko A, Chen S, Klebe D, Ma Q, Caner B, Fujii M, Zhou C, Zhang JH. Role of SCH79797 in maintaining vascular integrity in rat model of subarachnoid hemorrhage. Stroke 2013; 44:1410-7. [PMID: 23539525 DOI: 10.1161/strokeaha.113.678474] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND PURPOSE Plasma thrombin concentration is increased after subarachnoid hemorrhage (SAH). However, the role of thrombin receptor (protease-activated receptor-1 [PAR-1]) in endothelial barrier disruption has not been studied. The aims of this study were to investigate the role of PAR-1 in orchestrating vascular permeability and to assess the potential therapeutics of a PAR-1 antagonist, SCH79797, through maintaining vascular integrity. METHODS SCH79797 was injected intraperitoneally into male Sprauge-Dawley rats undergoing SAH by endovascular perforation. Assessment was conducted at 24 hours after SAH for brain water content, Evans blue content, and neurobehavioral testing. To explore the role of PAR-1 activation and the specific mechanism of SCH79797's effect after SAH, Western blot, immunoprecipitation, and immunofluorescence of hippocampus tissue were performed. A p21-activated kinase-1 (PAK1) inhibitor, IPA-3, was used to explore the underlying protective mechanism of SCH79797. RESULTS At 24 hours after SAH, animals treated with SCH79797 demonstrated a reduction in brain water content, Evans blue content, and neurobehavioral deficits. SCH79797 also attenuated PAR-1 expression and maintained the level of vascular endothelial-cadherin, an important component of adherens junctions. Downstream to PAR-1, c-Src-dependent activation of p21-activated kinase-1 led to an increased serine/threonine phosphorylation of vascular endothelial-cadherin; immunoprecipitation results revealed an enhanced binding of phosphorylated vascular endothelial-cadherin with endocytosis orchestrator β-arrestin-2. These pathological states were suppressed after SCH79797 treatment. CONCLUSIONS PAR-1 activation after SAH increases microvascular permeability, at least, partly through a PAR-1-c-Src-p21-activated kinase-1-vascular endothelial-cadherin phosphorylation pathway. Through suppressing PAR-1 activity, SCH79797 plays a protective role in maintaining microvascular integrity after SAH.
Collapse
Affiliation(s)
- Junhao Yan
- Department of Anatomy and Histology, School of Basic Medical Sciences, Peking University, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Dumas M, Nadal-Wollbold F, Gaussem P, Perez M, Mirault T, Létienne R, Bourbon T, Grelac F, Le Grand B, Bachelot-Loza C. Antiplatelet and antithrombotic effect of F 16618, a new thrombin proteinase-activated receptor-1 (PAR1) antagonist. Br J Pharmacol 2012; 165:1827-1835. [PMID: 21913899 DOI: 10.1111/j.1476-5381.2011.01668.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE New antithrombotic agents with the potential to prevent atherothrombotic complications are being developed to target receptors on platelets and other cells involved in plaque growth. The aim of this study was to investigate the antiplatelet effects of F 16618, a new non-peptidic PAR1 (thrombin receptor) antagonist. EXPERIMENTAL APPROACH We investigated the inhibitory effect of F 16618 on human platelet aggregation ex vivo, in whole blood and washed platelets, by using a multiple-electrode platelet aggregometer based on impedance and an optical aggregometer, respectively. Its effects on whole-blood haemostasis (clot parameters) were analysed with the ROTEM thromboelastometry device and the platelet function analyser PFA-100. A guinea-pig model of arterial thrombosis was used to investigate its effects on thrombus formation in vivo. KEY RESULTS F 16618 inhibited PAR1 agonist peptide (SFLLR-peptide)-induced washed platelet aggregation ex vivo. This effect was concentration-dependent and exhibited a competitive inhibition profile. Washed platelet aggregation, as well as P-selectin expression induced by thrombin, were significantly inhibited by 10 µM F 16618. In whole-blood experiments, 20 µM F 16618 inhibited SFLLR-induced platelet aggregation by 49%. In contrast, it had no effect on whole-blood haemostasis. In the guinea-pig model of carotid thrombosis, 0.32 mg·kg(-1) F 16618 doubled the occlusion time. CONCLUSIONS AND IMPLICATIONS F 16618 was shown to have strong antithrombotic activity in vivo and moderate antiplatelet effects ex vivo. As these effects were not associated with major effects on physiological haemostasis, this molecule is a good antiplatelet drug candidate for use either alone or in combination with current treatments.
Collapse
Affiliation(s)
- M Dumas
- Inserm UMR S 765, Faculté de Pharmacie, Paris, FranceUniversité Paris Descartes, Sorbonne Paris Cité, Paris, FranceCentre de Recherche Pierre Fabre, 17, Avenue Jean Moulin, Castres cedex, FranceAP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France
| | - F Nadal-Wollbold
- Inserm UMR S 765, Faculté de Pharmacie, Paris, FranceUniversité Paris Descartes, Sorbonne Paris Cité, Paris, FranceCentre de Recherche Pierre Fabre, 17, Avenue Jean Moulin, Castres cedex, FranceAP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France
| | - P Gaussem
- Inserm UMR S 765, Faculté de Pharmacie, Paris, FranceUniversité Paris Descartes, Sorbonne Paris Cité, Paris, FranceCentre de Recherche Pierre Fabre, 17, Avenue Jean Moulin, Castres cedex, FranceAP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France
| | - M Perez
- Inserm UMR S 765, Faculté de Pharmacie, Paris, FranceUniversité Paris Descartes, Sorbonne Paris Cité, Paris, FranceCentre de Recherche Pierre Fabre, 17, Avenue Jean Moulin, Castres cedex, FranceAP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France
| | - T Mirault
- Inserm UMR S 765, Faculté de Pharmacie, Paris, FranceUniversité Paris Descartes, Sorbonne Paris Cité, Paris, FranceCentre de Recherche Pierre Fabre, 17, Avenue Jean Moulin, Castres cedex, FranceAP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France
| | - R Létienne
- Inserm UMR S 765, Faculté de Pharmacie, Paris, FranceUniversité Paris Descartes, Sorbonne Paris Cité, Paris, FranceCentre de Recherche Pierre Fabre, 17, Avenue Jean Moulin, Castres cedex, FranceAP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France
| | - T Bourbon
- Inserm UMR S 765, Faculté de Pharmacie, Paris, FranceUniversité Paris Descartes, Sorbonne Paris Cité, Paris, FranceCentre de Recherche Pierre Fabre, 17, Avenue Jean Moulin, Castres cedex, FranceAP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France
| | - F Grelac
- Inserm UMR S 765, Faculté de Pharmacie, Paris, FranceUniversité Paris Descartes, Sorbonne Paris Cité, Paris, FranceCentre de Recherche Pierre Fabre, 17, Avenue Jean Moulin, Castres cedex, FranceAP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France
| | - B Le Grand
- Inserm UMR S 765, Faculté de Pharmacie, Paris, FranceUniversité Paris Descartes, Sorbonne Paris Cité, Paris, FranceCentre de Recherche Pierre Fabre, 17, Avenue Jean Moulin, Castres cedex, FranceAP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France
| | - C Bachelot-Loza
- Inserm UMR S 765, Faculté de Pharmacie, Paris, FranceUniversité Paris Descartes, Sorbonne Paris Cité, Paris, FranceCentre de Recherche Pierre Fabre, 17, Avenue Jean Moulin, Castres cedex, FranceAP-HP, Hôpital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France
| |
Collapse
|
22
|
Gadge ST, Khedkar MV, Lanke SR, Bhanage BM. Oxidative Aminocarbonylation of Terminal Alkynes for the Synthesis of Alk-2-ynamides by Using Palladium-on-Carbon as Efficient, Heterogeneous, Phosphine-Free, and Reusable Catalyst. Adv Synth Catal 2012. [DOI: 10.1002/adsc.201200041] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
23
|
Caliendo G, Santagada V, Perissutti E, Severino B, Fiorino F, Frecentese F, Juliano L. Kallikrein protease activated receptor (PAR) axis: an attractive target for drug development. J Med Chem 2012; 55:6669-86. [PMID: 22607152 DOI: 10.1021/jm300407t] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Giuseppe Caliendo
- Dipartimento di Chimica Farmaceutica e Tossicologica, Università degli Studi di Napoli Federico II, Via D. Montesano, 49, 80131, Napoli, Italy.
| | | | | | | | | | | | | |
Collapse
|
24
|
Lee H, Hamilton JR. Physiology, pharmacology, and therapeutic potential of protease-activated receptors in vascular disease. Pharmacol Ther 2012; 134:246-59. [DOI: 10.1016/j.pharmthera.2012.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 01/17/2012] [Indexed: 01/09/2023]
|
25
|
Dockendorff C, Aisiku O, VerPlank L, Dilks JR, Smith DA, Gunnink SF, Dowal L, Negri J, Palmer M, MacPherson L, Schreiber SL, Flaumenhaft R. Discovery of 1,3-Diaminobenzenes as Selective Inhibitors of Platelet Activation at the PAR1 Receptor. ACS Med Chem Lett 2012; 3:232-237. [PMID: 22408714 PMCID: PMC3297361 DOI: 10.1021/ml2002696] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 01/23/2012] [Indexed: 12/19/2022] Open
Abstract
![]()
A high-throughput screen of the NIH-MLSMR compound collection,
along with a series of secondary assays to identify potential targets
of hit compounds, previously identified a 1,3-diaminobenzene scaffold
that targets protease-activated receptor 1 (PAR1). We now report additional
structure–activity relationship (SAR) studies that delineate
the requirements for activity at PAR1 and identify plasma-stable analogues
with nanomolar inhibition of PAR1-mediated platelet activation. Compound 4 was declared as a probe (ML161) with the NIH Molecular Libraries
Program. This compound inhibited platelet aggregation induced by a
PAR1 peptide agonist or by thrombin but not by several other platelet
agonists. Initial studies suggest that ML161 is an allosteric inhibitor
of PAR1. These findings may be important for the discovery of antithrombotics
with an improved safety profile.
Collapse
Affiliation(s)
- Chris Dockendorff
- Chemical Biology Platform and
Probe Development Center, Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, Massachusetts
02142, United States
| | - Omozuanvbo Aisiku
- Division
of Hemostasis and Thrombosis,
Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215,
United States
| | - Lynn VerPlank
- Chemical Biology Platform and
Probe Development Center, Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, Massachusetts
02142, United States
| | - James R. Dilks
- Division
of Hemostasis and Thrombosis,
Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215,
United States
| | - Daniel A. Smith
- Division
of Hemostasis and Thrombosis,
Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215,
United States
| | - Susanna F. Gunnink
- Division
of Hemostasis and Thrombosis,
Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215,
United States
| | - Louisa Dowal
- Division
of Hemostasis and Thrombosis,
Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215,
United States
| | - Joseph Negri
- Chemical Biology Platform and
Probe Development Center, Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, Massachusetts
02142, United States
| | - Michelle Palmer
- Chemical Biology Platform and
Probe Development Center, Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, Massachusetts
02142, United States
| | - Lawrence MacPherson
- Chemical Biology Platform and
Probe Development Center, Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, Massachusetts
02142, United States
| | - Stuart L. Schreiber
- Chemical Biology Platform and
Probe Development Center, Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, Massachusetts
02142, United States
- Howard Hughes
Medical Institute, Broad Institute of Harvard and MIT, 7 Cambridge Center,
Cambridge, Massachusetts 02142, United States
| | - Robert Flaumenhaft
- Division
of Hemostasis and Thrombosis,
Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215,
United States
| |
Collapse
|
26
|
Targeting proteinase-activated receptors: therapeutic potential and challenges. Nat Rev Drug Discov 2012; 11:69-86. [PMID: 22212680 DOI: 10.1038/nrd3615] [Citation(s) in RCA: 247] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Proteinase-activated receptors (PARs), a family of four seven-transmembrane G protein-coupled receptors, act as targets for signalling by various proteolytic enzymes. PARs are characterized by a unique activation mechanism involving the proteolytic unmasking of a tethered ligand that stimulates the receptor. Given the emerging roles of these receptors in cancer as well as in disorders of the cardiovascular, musculoskeletal, gastrointestinal, respiratory and central nervous system, PARs have become attractive targets for the development of novel therapeutics. In this Review we summarize the mechanisms by which PARs modulate cell function and the roles they can have in physiology and diseases. Furthermore, we provide an overview of possible strategies for developing PAR antagonists.
Collapse
|
27
|
Chieng-Yane P, Bocquet A, Létienne R, Bourbon T, Sablayrolles S, Perez M, Hatem SN, Lompré AM, Le Grand B, David-Dufilho M. Protease-activated receptor-1 antagonist F 16618 reduces arterial restenosis by down-regulation of tumor necrosis factor α and matrix metalloproteinase 7 expression, migration, and proliferation of vascular smooth muscle cells. J Pharmacol Exp Ther 2011; 336:643-51. [PMID: 21139058 DOI: 10.1124/jpet.110.175182] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
Wound healing after angioplasty or stenting is associated with increased production of thrombin and the activation of protease-activated receptor 1 (PAR1). The aim of the present study was to examine the effects of a new selective PAR1 antagonist, 2-[5-oxo-5-(4-pyridin-2-ylpiperazin-1-yl)-penta-1,3-dienyl]-benzonitrile (F 16618), in restenosis and vascular smooth muscle cell (SMC) proliferation and migration using both in vivo and in vitro approaches. Daily oral administration of F 16618 inhibited the restenosis induced by balloon angioplasty on rat carotid artery in a dose-dependent manner. Furthermore, single intravenous administration of F 16618 during the angioplasty procedure was sufficient to protect the carotid artery against restenosis. In vitro, F 16618 inhibited the growth of human aortic SMCs in a concentration-dependent manner with maximal effects at 10 μM. At that concentration, F 16618 also prevented thrombin-mediated SMC migration. In vivo, oral and intravenous F 16618 treatments reduced by 30 and 50% the expression of the inflammatory cytokine tumor necrosis factor α (TNFα) 24 h after angioplasty. However, only acute intravenous administration prevented the induction of matrix metalloproteinase 7 expression. In contrast, F 16618 treatments had no effect on early SMC de-differentiation and transcription of monocyte chemoattractant protein-1 and interleukin-6 and late re-endothelialization of injured arteries. Furthermore, F 16618 compensated for the carotid endothelium loss by inhibiting PAR1-mediated contraction. Altogether, these data demonstrate that PAR1 antagonists such as F 16618 are a highly effective treatment of restenosis after vascular injury, by inhibition of TNFα, matrix metalloproteinase 7, and SMC migration and proliferation in addition to an antithrombotic effect.
Collapse
MESH Headings
- Animals
- Carotid Stenosis/metabolism
- Carotid Stenosis/prevention & control
- Cell Movement/drug effects
- Cell Movement/physiology
- Cell Proliferation/drug effects
- Cells, Cultured
- Coronary Restenosis/drug therapy
- Coronary Restenosis/metabolism
- Down-Regulation/drug effects
- Down-Regulation/physiology
- Gene Expression Regulation, Enzymologic
- Humans
- Male
- Matrix Metalloproteinase 7/biosynthesis
- Matrix Metalloproteinase Inhibitors
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/metabolism
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Pyridines/pharmacology
- Pyridines/therapeutic use
- Rats
- Rats, Sprague-Dawley
- Receptor, PAR-1/physiology
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/metabolism
- Platelet Aggregation Inhibitors
Collapse
|
28
|
Adams MN, Ramachandran R, Yau MK, Suen JY, Fairlie DP, Hollenberg MD, Hooper JD. Structure, function and pathophysiology of protease activated receptors. Pharmacol Ther 2011; 130:248-82. [PMID: 21277892 DOI: 10.1016/j.pharmthera.2011.01.003] [Citation(s) in RCA: 280] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 01/03/2011] [Indexed: 12/18/2022]
Abstract
Discovered in the 1990s, protease activated receptors(1) (PARs) are membrane-spanning cell surface proteins that belong to the G protein coupled receptor (GPCR) family. A defining feature of these receptors is their irreversible activation by proteases; mainly serine. Proteolytic agonists remove the PAR extracellular amino terminal pro-domain to expose a new amino terminus, or tethered ligand, that binds intramolecularly to induce intracellular signal transduction via a number of molecular pathways that regulate a variety of cellular responses. By these mechanisms PARs function as cell surface sensors of extracellular and cell surface associated proteases, contributing extensively to regulation of homeostasis, as well as to dysfunctional responses required for progression of a number of diseases. This review examines common and distinguishing structural features of PARs, mechanisms of receptor activation, trafficking and signal termination, and discusses the physiological and pathological roles of these receptors and emerging approaches for modulating PAR-mediated signaling in disease.
Collapse
Affiliation(s)
- Mark N Adams
- Mater Medical Research Institute, Aubigny Place, Raymond Terrace, South Brisbane Qld 4101, Australia
| | | | | | | | | | | | | |
Collapse
|
29
|
Cirino G, Severino B. Thrombin receptors and their antagonists: an update on the patent literature. Expert Opin Ther Pat 2010; 20:875-84. [PMID: 20450349 DOI: 10.1517/13543776.2010.487864] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
IMPORTANCE OF THE FIELD Thrombin plays a central role in cardiovascular inflammation. Most of the cellular responses to thrombin are mediated by cell surface protease-activated receptors (PARs). Several preclinical studies indicate that PARs are potential targets for treating cardiovascular diseases such as thrombosis, atherosclerosis and restenosis. Among PARs, PAR-1 has emerged as an important therapeutic target. AREAS COVERED IN THIS REVIEW This review covers recent advances in the development of thrombin receptors antagonists. It is focused on the search for PAR-1 antagonists as this is at the moment the most promising and attractive target. However, some early promising studies on PAR-3 and -4 antagonists are also reported. WHAT THE READER WILL GAIN The review has been written in order to give to the reader hints and references that cover, in our opinion, the most interesting and/or promising approaches in this research field. TAKE HOME MESSAGE Research on PAR-1 antagonists has finally led to good clinical candidates such as SCH-530348 (Schering-Plough) and E-5555 (Eisai Co.). Clinical trials clearly demonstrate that development of PAR1 antagonists is not only possible but most likely will lead to development of antiplatelet drugs as well as of drugs useful for the treatment of inflammatory, proliferative and neurodegenerative diseases.
Collapse
Affiliation(s)
- Giuseppe Cirino
- University of Naples Federico II, Department of Experimental Pharmacology, Via Domenico Montesano 49, Napoli 80131, Italy.
| | | |
Collapse
|
30
|
Létienne R, Leparq-Panissié A, Calmettes Y, Nadal-Wollbold F, Perez M, Le Grand B. Antithrombotic activity of F 16618, a new PAR1 antagonist evaluated in extracorporeal arterio-venous shunt in the rat. Biochem Pharmacol 2010; 79:1616-21. [DOI: 10.1016/j.bcp.2010.02.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 02/04/2010] [Accepted: 02/08/2010] [Indexed: 11/26/2022]
|
31
|
Planty B, Pujol C, Lamothe M, Maraval C, Horn C, Grand BL, Perez M. Exploration of a new series of PAR1 antagonists. Bioorg Med Chem Lett 2010; 20:1735-9. [DOI: 10.1016/j.bmcl.2010.01.050] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 01/06/2010] [Accepted: 01/07/2010] [Indexed: 11/17/2022]
|
32
|
Rilatt I, Mirabel E, Grand BL, Perez M. Discovery and SAR of small molecule PAR1 antagonists. Bioorg Med Chem Lett 2010; 20:903-6. [DOI: 10.1016/j.bmcl.2009.12.072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 12/16/2009] [Accepted: 12/17/2009] [Indexed: 10/20/2022]
|