Volume : 10, Issue : 01, January – 2023

Title:

01.REVIEW ON: “COUMARINS: A Unique Scaffold with Versatile Biological Behaviour”

Authors :

*Vaishnavi U. Kale, Vaishnavi S. Balinge, Dr. Anjali M. Wankhade, Nikhil S. Wagh,

Abstract :

The Tonka bean, from which coumarin was first isolated in 1820, is represented by the word “coumarins,” which also refers to “coumarou.” A natural substance obtained from plants called coumarin (2H-1-benzopyran-2-one) is well known for its pharmacological effects, including antibacterial, antifungal, antiviral, antitubercular, anticancer, antihypertensive characteristics that are anti-adipogenic, anti-hyperglycemic, antioxidant, and neuroprotective. dietary contact with Benzopyrones are significant since they are present in many foods, including seeds, nuts, coffee, and vegetables, wine and tea. Considering the known low toxicity, relative affordability, availability in the diet, and Given that coumarins are present in many herbal treatments, it would be wise to consider their characteristics and other applications. It consists of categorization and pharmacologic factors like absorption.
Keywords: Coumarin, Tonka Beans, Benzopyrene, Toxicological Studies, Antiadipogenic

Cite This Article:

Please cite this article in press Vaishnavi U. Kale et al, Review on: “coumarins: a unique scaffold with versatile biological behaviour”., Indo Am. J. P. Sci, 2023; 10(01).

Number of Downloads : 10

References:

1. Sarker SD, Nahar L. Progress in the chemistry of naturally occurring coumarins. ProgChem Org Nat Prod 2017;106:241-304.
2. Pangal A, Gazge M, Mane V, Shaikh JA. Various pharmacological aspects of couamrin derivatives: A review. Int J Pharm Res Biosci 2013;2:168-94.
3. Rohini K, Srikumar PS. Therapeutic role of coumarins and coumarinrelated compounds. J ThermodynCatal 2014;5:1-3.
4. Constantinou AI, Kamath N, Murley JS. Genistein inactivates bcl-2, delays the G2/M phase of the cell cycle, and induces apoptosis of human breast adenocarcinoma MCF-7 cells. Eur J Cancer 1998;34:1927-34.
5. Egan D, O’Kennedy R, Moran E, Cox D, Prosser E, Thornes RD, et al. The pharmacology, metabolism, analysis, and applications of coumarin and coumarin-related compounds. Drug Metab Rev 1990;22:503-29.
6. Egan DA, O’Kennedy R. Rapid and sensitive determination of coumarin and 7-hydroxycoumarin and its glucuronide conjugate in urine and plasma by high-performance liquid chromatography. J Chromatogr 1992;582:137-43.
7. Finn GJ, Kenealy E, Creaven BS, Egan DA. In vitro cytotoxic potential and mechanism of action of selected coumarins, using human renal cell lines. Cancer Lett 2002;183:61-8.
8. Ritschel WA, Grummich KW, Kaul S, Hardt TJ. Biopharmaceutical parameters of coumarin and 7-hydroxycoumarin. Die PharmaInd 1981;43:271-6.
9. Stiefel C, Schubert T, Morlock GE. Bioprofiling of cosmetics with focus and streamlined coumarin analysis. ACS Omega 2017;2:5242-50.
10. Dempsey E, O’Sullivan C, Smyth MR, Egan D, O’Kennedy R, Wang J, et al. Development of an antibody-based amperometric biosensor to study the reaction of 7-hydroxycoumarin with its specific antibody. Analyst 1993;118:411-3.
11. Sharma S, Byrne H, O’Kennedy RJ. Antibodies and antibody-derived analytical biosensors. Essays Biochem 2016;60:9-18.
12. Shilling WH, Crampton RF, Longland RC. Metabolism of coumarin in man. Nature 1969;221:664-5.
13. Moran E, O’Kennedy R, Thornes RD. Analysis of coumarin and its urinary metabolites by high-performance liquid chromatography. J Chromatogr 1987;416:165-9.
14. Pelkonen O, Turpeinen M, Uusitalo J, Rautio A, Raunio H. Prediction of drug metabolism and interactions on the basis of in vitro investigations. Basic ClinPharmacolToxicol 2005;96:167-75.
15. Bogan DP, Deasy B, O’Kennedy R, Smyth MR, Fuhr U. Determination of free and total 7-hydroxycoumarin in urine and serum by capillary electrophoresis. J Chromatogr B Biomed Appl 1995;663:371-8.
16. Rautio A, Kraul H, Kojo A, Salmela E, Pelkonen O. Interindividual variability of coumarin 7-hydroxylation in healthy volunteers. Pharmacogenetics 1992;2:227-33.
17. Reddy DS, Kongot M, Netalkar SP, Kurjogi MM, Kumar R, Avecilla F, et al. Synthesis and evaluation of novel coumarin-oxime ethers as potential anti-tubercular agents: Their DNA cleavage ability and BSA interaction study. Eur J Med Chem 2018;150:864-75.
18. Mangasuli SN, Hosamani KM, Devarajegowda HC, Kurjogi MM, JoshiSD. Synthesis of coumarin-theophylline hybrids as a new class of anti-tubercular and anti-microbial agents. Eur J Med Chem 2018;146:747-56.
19. Benci K, Mandić L, Suhina T, Sedić M, Klobučar M, KraljevićPavelić S, et al. Novel coumarin derivatives containing 1,2,4-triazole, 4,5-dicyanoimidazole and purine moieties: Synthesis and evaluation of their cytostatic activity. Molecules 2012;17:11010-25.
20. Cheng JF, Chen M, Wallace D, Tith S, Arrhenius T, Kashiwagi H, et al. Discovery and structure-activity relationship of coumarin derivatives as TNF-alpha inhibitors. Bioorg Med ChemLett 2004;14:2411-5.
21. Lee S, Sivakumar K, Shin WS, Xie F, Wang Q. Synthesis and antiangiogenesis activity of coumarin derivatives. Bioorg Med ChemLett 2006;16:4596-9.
22. Chen W, Tang W, Lou L, Zhao W. Pregnane, coumarin and lupane derivatives and cytotoxic constituents from Helicteresangustifolia. Phytochemistry 2006;67:1041-7.
23. Nofal ZM, El-Zahar MI, El-Karim SS. Novel coumarin derivatives with expected biological activity. Molecules 2000;5:99-113.
24. Lou LL, Zhao P, Cheng ZY, Guo R, Yao GD, Wang XB, et al. A new coumarin from Juglansmandshurica maxim induce apoptosis in hepatocarcinoma cells. Nat Prod Res 2018;1-3. Avaliable from: 10.1080/14786419.2018.1434646 [Last accessed on 2018 Feb 04].
25. Tan S, He F, Kong T, Wu J, Liu Z. Design, synthesis and tumor cell growth inhibitory activity of 3-nitro-2H-cheromene derivatives as histone deacetylaes inhibitors. Bioorg Med Chem 2017;25:4123-32.
26. Lv N, Sun M, Liu C, Li J. Design and synthesis of 2-phenylpyrimidine coumarin derivatives as anticancer agents. Bioorg Med ChemLett 2017;27:4578-81.
27. Yu H, Hou Z, Tian Y, Mou Y, Guo C. Design, synthesis, cytotoxicity and mechanism of novel dihydroartemisinin-coumarin hybrids as potential anti-cancer agents. Eur J Med Chem 2018;151:434-49.
28. Fentem JH, Fry JR. Metabolism of coumarin by rat, gerbil and human liver microsomes. Xenobiotica 1992;22:357-67.
29. Lin PY, Yeh KS, Su CL, Sheu SY, Chen T, Ou KL, et al. Synthesis and antibacterial activities of novel 4-hydroxy-7-hydroxy- and 3-carboxycoumarin derivatives. Molecules 2012;17:10846-63.
30. Hu Y, Shen Y, Wu X, Tu X, Wang GX. Synthesis and biological evaluation of coumarin derivatives containing imidazole skeleton as potential antibacterial agents. Eur J Med Chem 2018;143:958-69.
31. Amin KM, Abou-Seri SM, Abdelnaby RM, Rateb HS, Khalil MA, Hussein MM. Synthesis and biological evaluation of novel coumarin derivatives as potential antimicrobial agents. Int J Pharm PharmSci 2016;8:109-19.
32. Aziem AA. An efficient and simple synthesis of 2, 3-dihydro-1,3,4- thiazoles, pyrazoles and coumarin containing benzofuran moiety using both conventional and grinding methods. Int J Pharm PharmSci 2015;7:61-8.
33. Hamid SJ, Kubba AA. Synthesis and characterization of new coumarin derivatives containing various moieties with antibacterial activities. Int J Pharm PharmSci 2015;7:70-4.
34. Tandon R, Ponnan P, Aggarwal N, Pathak R, Baghel AS, Gupta G, et al. Characterization of 7-amino-4-methylcoumarin as an effective antitubercular agent: Structure-activity relationships. J AntimicrobChemother 2011;66:2543-55.
35. Singh H, Singh JV, Gupta MK, Saxena AK, Sharma S, Nepali K, et al. Triazole tethered isatin-coumarin based molecular hybrids as novel antitubulin agents: Design, synthesis, biological investigation and docking studies. Bioorg Med ChemLett 2017;27:3974-9.
36. Tiwari SV, Seijas JA, Vazquez-Tato MP, Sarkate AP, Karnik KS, Nikalje APG, et al. Facile synthesis of novel coumarin derivatives, antimicrobial analysis, enzyme assay, docking study, ADMET prediction and toxicity study. Molecules 2017;22:E1172.
37. Bu M, Cao T, Li H, Guo M, Yang BB, Zeng C, et al. Synthesis and biological evaluation of novel steroidal 5α,8α-epidioxyandrost-6-ene3β-ol-17-(O-phenylacetamide)oxime derivatives as potential anticancer agents. Bioorg Med ChemLett 2017;27:3856-61.