1
|
Chen L, Lv Q, Cai J, Liang J, Liang Z, Lin J, Xiao Y, Chen R, Zhang Z, Hong Y, Ji H. Design, synthesis and anticancer activity studies of 3-(coumarin-3-yl)-acrolein derivatives: Evidenced by integrating network pharmacology and vitro assay. Front Pharmacol 2023; 14:1141121. [PMID: 37033621 PMCID: PMC10076643 DOI: 10.3389/fphar.2023.1141121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Coumarin derivatives have diverse structures and show various significant biological activities. Aiming to develop more potent coumarin derivatives for cancer treatment, a series of coumarin acrolein hybrids were designed and synthesized by using molecular hybridization approach, and investigated for their antiproliferative activity against A549, KB, Hela and MCF-7 cancer cells as well as HUVEC and LO2 human normal cells. The results indicated that most of the synthesized compounds displayed remarkable inhibitory activity towards cancer cells but low cytotoxicity on normal cells. Among all the compounds, 5d and 6e were the most promising compounds against different cancer cell lines, especially for A549 and KB cells. The preliminary action mechanism studies suggested that compound 6e, the representative compound, was capable of dose-dependently suppressing migration, invasion and inducing significant apoptosis. Furthermore, the combined results of network pharmacology and validation experiments revealed that compound 6e induced mitochondria dependent apoptosis via the PI3K/AKT-mediated Bcl-2 signaling pathway. In summary, our study indicated compound 6e could inhibit cell proliferation, migration, invasion and promote cell apoptosis through inhibition of PI3K/AKT signaling pathway in human oral epidermoid carcinoma cells. These findings demonstrated the potential of 3-(coumarin-3-yl)-acrolein derivatives as novel anticancer chemotherapeutic candidates, providing ideas for further development of drugs for clinical use.
Collapse
Affiliation(s)
- Lexian Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qianqian Lv
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jianghong Cai
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Taipa, China
| | - Jiajie Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ziyan Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiahui Lin
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ying Xiao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ruiyao Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhiling Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yue Hong
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Hong Ji
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Hong Ji,
| |
Collapse
|
2
|
Sinha S, Singh K, Ved A, Hasan SM, Mujeeb S. Therapeutic Journey and Recent Advances in the Synthesis of Coumarin Derivatives. Mini Rev Med Chem 2021; 22:1314-1330. [PMID: 34784861 DOI: 10.2174/1389557521666211116120823] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/27/2021] [Accepted: 09/03/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Coumarin is an oxygen-containing compound in medicinal chemistry. Coumarin plays an important role in both natural systems like plants and also in synthetic medicinal applications as drug molecules. Many structurally different coumarin compounds were found to show a big range of similarity with the vital molecular targets for their pharmacological action and small modifications in their structures resulted insignificant changes in their biological activities. OBJECTIVE This review gives detailed information about the studies of the recent advances in various pharmacological aspects of coumarins. METHOD Various oxygen-containing heterocyclic compounds represented remarkable biological significances. The fused aromatic oxygen-heterocyclic nucleus is able to change its electron density; thus changing the chemical, physical and biological properties respectively due to its multiple binding modes with the receptors, which play crucial role in pharmacological screening of drugs. A number of heterocyclic compounds have been synthesized which have their nucleus derived from various plants and animals. In coumarins, benzene ring is fused with pyrone nucleus which provides stability to the nucleus. Coumarins have shown a wide range of pharmacological activities such as anti-tumour, anti-coagulant, anti-inflammatory, anti-oxidant, antiviral, anti-malarial, anti-HIV and antimicrobial activity etc. Results: Reactive oxygen species like superoxide anion, hydroxyl radical and hydrogen peroxide are a type of unstable molecule that contains oxygen, which reacts with other molecules in the cell during the metabolism process but it may produce cytotoxicity when reactive oxygen species increase in number, by the damage of biological macromolecules. Hydroxyl radical (˙OH), is a strong oxidizing agent and it is responsible for the cytotoxicity by oxygen in different plants, animals and other microbes. coumarin is the oldest and effective compound having antimicrobial activity, anti-inflammatory, antioxidant, antidepressant activity, analgesic, anticonvulsant activity, etc. Naturally existing coumarin compounds act against SARS-CoV-2 by preventing viral replication through the targeting on active site against the Mpro target protein. CONCLUSION This review highlights the different biological activities of coumarin derivatives. In this review we provide an updated summary of the researches which are related to recent advances in biological activities of coumarins analogue and their most recent activities against COVID -19. Natural compounds act as a rich resource for novel drug development against various SARS-CoV-2 viral strains including viruses like herpes simplex virus, influenza virus, human immunodeficiency virus, hepatitis B and C viruses, middle east respiratory syndrome and severe acute respiratory syndrome.
Collapse
Affiliation(s)
- Shweta Sinha
- Faculty of Pharmacy, Integral University, Kursi road, Lucknow-226026 (U.P.). India
| | - Kuldeep Singh
- Faculty of Pharmacy, Integral University, Kursi road, Lucknow-226026 (U.P.). India
| | - Akash Ved
- Goel Institute of Pharmaceutical Sciences, Lucknow -226028 (U.P.). India
| | - Syed Misbahul Hasan
- Faculty of Pharmacy, Integral University, Kursi road, Lucknow-226026 (U.P.). India
| | - Samar Mujeeb
- Hygia Institute of Pharmaceutical Education and Research, Lucknow. India
| |
Collapse
|
3
|
The antithrombotic activity of natural and synthetic coumarins. Fitoterapia 2021; 154:104947. [PMID: 34352355 DOI: 10.1016/j.fitote.2021.104947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/29/2021] [Accepted: 05/30/2021] [Indexed: 12/30/2022]
Abstract
Thrombosis, which seriously endangers human health and life, is the leading cause of morbidity and mortality globally. Antithrombotic drugs can interfere with the occurrence and development of thrombotic diseases and play an important role in the treatment of thrombotic diseases. However, unsatisfactory efficacy and serious adverse effects of existing antithrombotic drugs increase the research for new, efficient and safer drugs. Natural and synthetic coumarins have been shown to possess antithrombotic activity, namely, anticoagulation and antiplatelet aggregation. Especially, coumarin-based warfarin, phenprocoumon and cloricromen have long been used in clinical treatment of thrombosis. Coumarin with low toxicity is the privileged structure for developing novel antithrombotic drugs with multiple mechanisms of action. The present review aims to compile current research on the development of coumarins against thrombosis, emphasizing the relationship between their chemical structures and therapeutic effectiveness. It is intended to provide promising ideas for the discovery of novel coumarin derivatives with high antithrombotic activity.
Collapse
|
4
|
Singh LR, Chen YL, Xie YY, Xia W, Gong XW, Hider RC, Zhou T. Functionality study of chalcone-hydroxypyridinone hybrids as tyrosinase inhibitors and influence on anti-tyrosinase activity. J Enzyme Inhib Med Chem 2021; 35:1562-1567. [PMID: 32746652 PMCID: PMC7470021 DOI: 10.1080/14756366.2020.1801669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In an attempt to synthesise new tyrosinase inhibitors, we designed and synthesised a series of chalcone-hydroxypyridinone hybrids as potential tyrosinase inhibitors adopting strategic modifications of kojic acid. All the newly synthesised compounds were characterised by NMR and mass spectrometry. Initial screening of the target compounds demonstrated that compounds 1a, 1d, and 1n had relatively strong inhibitory activities against tyrosinase monophenolase, with IC50 values of 3.07 ± 0.85, 2.25 ± 0.8 and 2.75 ± 1.19 μM, respectively. The inhibitory activity against monophenolase was 6- to 8-fold higher than that of kojic acid. Compounds 1a, 1d, and 1n also showed inhibition of diphenolase, with IC50 values of 17.05 ± 0.07, 11.70 ± 0.03 and 19.3 ± 0.28 μM, respectively. The inhibition kinetics of diphenolase indicates that compounds 1a and 1d induce reversible inhibition on tyrosinase. Finally, we found that copper coordination should be one of the important inhibitory mechanism of these compounds in tyrosinase.
Collapse
Affiliation(s)
- L Ravithej Singh
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, PR China
| | - Yu-Lin Chen
- Division of Pharmaceutical Science, King's College London, London, UK
| | - Yuan-Yuan Xie
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, PR China
| | - Wei Xia
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, PR China
| | - Xing-Wen Gong
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, PR China
| | - Robert C Hider
- Division of Pharmaceutical Science, King's College London, London, UK
| | - Tao Zhou
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, PR China
| |
Collapse
|
5
|
ZnCl 2 catalyzed new coumarinyl-chalcones as cytotoxic agents. Saudi J Biol Sci 2021; 28:386-394. [PMID: 33424321 PMCID: PMC7783654 DOI: 10.1016/j.sjbs.2020.10.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 11/27/2022] Open
Abstract
A new series of coumarin-yl-chalcone derivatives (3a-m) had been designed and synthesized through different reactions such as aromatic addition, cyclization and Claisen-Schmidt reactions in good yields (54–78%). 5-acetyl-4-(2-hydroxyphenyl) -6-methyl-3, 4-dihydropyrimidin-2(1H) -one (1) has been synthesized by multi-component one pot reaction of salicylaldehyde, methyl acetoacetate and urea, which was further reacted with malonic acid employing ZnCl2 catalyst to yield 5-acetyl-4-(4-hydroxy-2-oxo-2H-chromen-8-yl) -6-methyl-3, 4-dihydropyrimidin-2(1H) -one (2). The title compounds (3a-m) were synthesised by reacting 5-acetyl-4-(4-hydroxy-2-oxo-2H-chromen-8-yl) -6-methyl-3, 4-dihydropyrimidin-2(1H)-one (2) with different aromatic aldehydes in the presence of potassium hydroxide. In silico studies, a preliminary screening method for predicting the anti-cancer activity was performed for the synthesized compounds (3a-m) against Src, Alb tyrosine kinase and homology model protein (PDB ID: 4csv). The derivatives 3h and 3m showed moderate binding energies. The in vitro cytotoxic activity was evaluated for the compounds 3h and 3m by using human cancer cell-line morphology and MTT assay against three human cell-lines A549 (Lung), Jurkat (Leukemia) and MCF-7 (Breast). The results indicate that the derivatives 3h and 3m display significant anti-cancer activity, however it was found to be less cytotoxic when compared to the standard used i.e. Imatinib.
Collapse
|
6
|
Perrone MG, Miciaccia M, Vitale P, Ferorelli S, Araújo CDCB, de Almeida GS, Souza Domingos TF, da Silva LCRP, de Pádula M, Cabral LM, Sathler PC, Bonaccorso C, Fortuna CG, Scilimati A. An attempt to chemically state the cross-talk between monomers of COX homodimers by double/hybrid inhibitors mofezolac-spacer-mofezolac and mofezolac-spacer-arachidonic acid. Eur J Med Chem 2020; 209:112919. [PMID: 33129592 DOI: 10.1016/j.ejmech.2020.112919] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/18/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases (CVDs) account for over 17 million death globally each year, including arterial thrombosis. Platelets are key components in the pathogenesis of this disease and modulating their activity is an effective strategy to treat such thrombotic events. Cyclooxygenase-1 (COX-1) isoenzyme is involved in platelet activation and is the main target of non-steroidal anti-inflammatory drugs (NSAIDs) and new selective inhibitor research. Inhibitors of general formula mofezolac-spacer-mofezolac (mof-spacer-mof) and mofezolac-spacer-arachidonic acid (mof-spacer-AA) were projected to investigate the possible cross-talk between the two monomers (Eallo and Ecat) forming the COX-1 homodimer. Mofezolac was chosen as either one or two moieties of these molecules being the known most potent and selective COX-1 inhibitor and administrated to humans as Disopain™, then arachidonic acid (AA) was used to develop molecules bearing, in the same compound, in addition to the inhibitor moiety (mofezolac) also the natural COX substrate. Depending on the nature of the spacer, COX-1 and COX-2 activity was differently inhibited by mof-spacer-mof set with a preferential COX-1 inhibition. The highest COX-1 selectivity was exhibited by the compound in which the spacer was the benzidine [N,N'-(biphenyl-4,4'-di-yl)bis (2-[3,4-bis(4-methoxyphenyl)isoxazol-5-yl]acetamide) (15): COX-1 IC50 = 0.08 μM, COX-2 IC50 > 50 μM, Selectivity Index (SI) > 625]. In the case of mof-spacer-AA set, the COX inhibitory potency and also the isoform preference changed. (5Z, 8Z, 11Z, 14Z)-N-(4-{2-[3,4-Bis(4-methoxyphenyl)isoxazol-5-yl]acetamido}butyl)icosa-5,8,11,14-tetraenamide (19) and (5Z, 8Z, 11Z, 14Z)-N-(4'-{2-[3,4-bis(4-methoxyphenyl)isoxazol-5-yl]acetamido}-[1,1'-biphenyl]-4-yl)icosa-5,8,11,14-tetraenamide (21), in which the spacer is the 1,2-diaminobutane or benzidine, respectively, selectively inhibited the COX-2, whereas when the spacer is the 1,4-phenylendiamine [(5Z, 8Z, 11Z, 14Z)-N-(4-{2-[3,4-bis(4-methoxyphenyl)isoxazol-5-yl]acetamido}phenyl)icosa-5,8,11,14-tetraenamide) (20) the COX preference is COX-1 (COX-1 IC50 = 0.05 μM, COX-2 IC50 > 50 μM, with a COX-1 selectivity > 1000). Molecular modelling by using FLAP algorithm shows fundamental interactions of the novel compounds at the entry channel of COX and inside its catalytic cavity. The effect of these mof-spacer-mof and mof-spacer-AA in inhibiting in vitro free arachidonic acid-induced platelet aggregation was also determined. A positive profile of hemocompatibility in relation to their influence on the blood coagulation cascade and erythrocyte toxicity was observed. Cytotoxicity and genotoxicity safety were also found for these two novel sets of compounds.
Collapse
Affiliation(s)
- Maria Grazia Perrone
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy
| | - Morena Miciaccia
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy
| | - Paola Vitale
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy
| | - Savina Ferorelli
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy
| | - Cristina da Costa Bernardes Araújo
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Gabriella Silva de Almeida
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Thaisa Francielle Souza Domingos
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | | | - Marcelo de Pádula
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Lucio Mendes Cabral
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Plínio Cunha Sathler
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Center of Health Sciences, Carlos Chagas Filho Avenue, 373, 21941599, Rio de Janeiro, Brazil
| | - Carmela Bonaccorso
- Department of Chemical Science, University of Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Cosimo G Fortuna
- Department of Chemical Science, University of Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Antonio Scilimati
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Via E. Orabona 4, 70125, Bari, Italy.
| |
Collapse
|
7
|
Nabeel A. Abdul-Rida, Adnan S, Jaber QAH. Development of Novel Imaging Fluorescent Agents Bearing Anti-Inflammatory Drugs: Synthesis, Structural Characterization and Evaluation of Biological Activity. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2020. [DOI: 10.1134/s1068162020040032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
8
|
Pati ML, Vitale P, Ferorelli S, Iaselli M, Miciaccia M, Boccarelli A, Di Mauro GD, Fortuna CG, Souza Domingos TF, Rodrigues Pereira da Silva LC, de Pádula M, Cabral LM, Sathler PC, Vacca A, Scilimati A, Perrone MG. Translational impact of novel widely pharmacological characterized mofezolac-derived COX-1 inhibitors combined with bortezomib on human multiple myeloma cell lines viability. Eur J Med Chem 2019; 164:59-76. [DOI: 10.1016/j.ejmech.2018.12.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 01/07/2023]
|
9
|
Sashidhara KV, Rao KB, Sonkar R, Modukuri RK, Chhonker YS, Kushwaha P, Chandasana H, Khanna AK, Bhatta RS, Bhatia G, Suthar MK, Saxena JK, Kumar V, Siddiqi MI. Hybrids of coumarin–indole: design, synthesis and biological evaluation in Triton WR-1339 and high-fat diet induced hyperlipidemic rat models. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00283h] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Lipid lowering activity of novel coumarin–indole hybrids has been demonstrated.
Collapse
|
10
|
Lourenço AL, Saito MS, Dorneles LEG, Viana GM, Sathler PC, Aguiar LCDS, de Pádula M, Domingos TFS, Fraga AGM, Rodrigues CR, de Sousa VP, Castro HC, Cabral LM. Synthesis and antiplatelet activity of antithrombotic thiourea compounds: biological and structure-activity relationship studies. Molecules 2015; 20:7174-200. [PMID: 25903367 PMCID: PMC6272548 DOI: 10.3390/molecules20047174] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 04/08/2015] [Accepted: 04/13/2015] [Indexed: 12/31/2022] Open
Abstract
The incidence of hematological disorders has increased steadily in Western countries despite the advances in drug development. The high expression of the multi-resistance protein 4 in patients with transitory aspirin resistance, points to the importance of finding new molecules, including those that are not affected by these proteins. In this work, we describe the synthesis and biological evaluation of a series of N,N'-disubstituted thioureas derivatives using in vitro and in silico approaches. New designed compounds inhibit the arachidonic acid pathway in human platelets. The most active thioureas (compounds 3d, 3i, 3m and 3p) displayed IC50 values ranging from 29 to 84 µM with direct influence over in vitro PGE2 and TXA2 formation. In silico evaluation of these compounds suggests that direct blockage of the tyrosyl-radical at the COX-1 active site is achieved by strong hydrophobic contacts as well as electrostatic interactions. A low toxicity profile of this series was observed through hemolytic, genotoxic and mutagenic assays. The most active thioureas were able to reduce both PGE2 and TXB2 production in human platelets, suggesting a direct inhibition of COX-1. These results reinforce their promising profile as lead antiplatelet agents for further in vivo experimental investigations.
Collapse
Affiliation(s)
- André Luiz Lourenço
- Programa de Pós-graduação em Patologia, Departamento de Patologia, Hospital Universitário Antônio Pedro (HUAP), Universidade Federal Fluminense (UFF), Niterói CEP 24033-900, RJ, Brazil.
| | - Max Seidy Saito
- Programa de Pós-graduação em Patologia, Departamento de Patologia, Hospital Universitário Antônio Pedro (HUAP), Universidade Federal Fluminense (UFF), Niterói CEP 24033-900, RJ, Brazil.
| | - Luís Eduardo Gomes Dorneles
- LabTIF, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro CEP 21941-902, RJ, Brazil.
| | - Gil Mendes Viana
- LabTIF, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro CEP 21941-902, RJ, Brazil.
| | - Plínio Cunha Sathler
- LabTIF, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro CEP 21941-902, RJ, Brazil.
| | | | - Marcelo de Pádula
- LabTIF, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro CEP 21941-902, RJ, Brazil.
| | | | - Aline Guerra Manssour Fraga
- LabTIF, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro CEP 21941-902, RJ, Brazil.
| | - Carlos Rangel Rodrigues
- ModMolQSAR, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro CEP 21941-902, RJ, Brazil.
| | - Valeria Pereira de Sousa
- LabTIF, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro CEP 21941-902, RJ, Brazil.
| | - Helena Carla Castro
- LABiEMOL, Departamento de Biologia Celular e Molecular, Universidade Federal Fluminense (UFF), Niterói CEP 24033-900, RJ, Brazil.
| | - Lucio Mendes Cabral
- LabTIF, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro CEP 21941-902, RJ, Brazil.
| |
Collapse
|
11
|
Khandelwal K, Pachauri SD, Arya A, Pawar VK, Joshi T, Dwivedi P, Ahmad H, Singh B, Sharma K, Kanojiya S, Chourasia MK, Saxena AK, Dwivedi AK. Improved oral bioavailability of novel antithrombotic S002-333 via chitosan coated liposomes: a pharmacokinetic assessment. RSC Adv 2015. [DOI: 10.1039/c5ra01543j] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
S002-333, a novel anti-thrombotic agent, exhibits excellent platelet mediated antithrombotic action and subsequently has no effect on the coagulation cascade.
Collapse
Affiliation(s)
- Kiran Khandelwal
- Pharmaceutics Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
- Jawaharlal Nehru University
| | | | - Abhishek Arya
- Pharmaceutics Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
- Academy of Scientific & Innovative Research
| | - Vivek K. Pawar
- Pharmaceutics Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
- Academy of Scientific & Innovative Research
| | - Trapti Joshi
- SAIF Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
- Jawaharlal Nehru University
| | - Pankaj Dwivedi
- Pharmaceutics Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
| | - Hafsa Ahmad
- Pharmaceutics Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
| | - Bupendra Singh
- Pharmaceutics Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
| | - Komal Sharma
- Pharmaceutics Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
- Academy of Scientific & Innovative Research
| | | | | | - Anil Kumar Saxena
- Medicinal and Process Chemistry Division
- CSIR-Central Drug Research Institute
- Lucknow
- India
| | | |
Collapse
|
12
|
Sathler PC, Lourenço AL, Rodrigues CR, da Silva LCRP, Cabral LM, Jordão AK, Cunha AC, Vieira MCB, Ferreira VF, Carvalho-Pinto CE, Kang HC, Castro HC. In vitro and in vivo analysis of the antithrombotic and toxicological profile of new antiplatelets N-acylhydrazone derivatives and development of nanosystems. Thromb Res 2014; 134:376-83. [DOI: 10.1016/j.thromres.2014.05.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 04/04/2014] [Accepted: 05/06/2014] [Indexed: 11/24/2022]
|