Main Article Content

Abstract

Nowadays, leukotriene antagonistic agents are playing an important role in the management of asthma, rhinitis and other inflammatory diseases of the lower respiratory tract. Leukotriene antagonistic agents available in the market are montelukast, pranulukast, zafirlukast, iralukast, cinalukast, zileuton, verlukst and so on. However, due to several side effects of above allopathic medicines, the bioactive compounds of plant origin are playing a very important role as secondary metabolites. These bioactive compounds are still being used by the human beings since time immemorial in the form of herbal preparations for the treatment of various ailments as mentioned in Ayurvedic system of medicine (ASM). Many researches have been reported related to the anti-inflammatory properties of plants in the traditional medicines, which are capable of suppressing, reducing and relieving pain as well as in reducing inflammation. Therefore, there is a need to highlights some plant species and their by-products possessing anti-inflammatory and leukotriene antagonistic properties.

Keywords

Phytochemicals Antagonistic agents Anti-inflammatory Leukotriene.

Article Details

How to Cite
Bhardwaj, K., Meshram, D., & Kumar Soni, K. (2020). A Review on Leukotriene Antagonistic Agents of Plant Origin. Asian Journal of Organic & Medicinal Chemistry, 5(1), 30–35. https://doi.org/10.14233/ajomc.2020.AJOMC-P241

References

  1. S. Hammarstrom, Leukotrienes, Annu. Rev. Biochem., 52, 355 (1983); https://doi.org/10.1146/annurev.bi.52.070183.002035
  2. J.N. Sharma and L.A. Mohammed, The Role of Leukotrienes in the Pathophysiology of Inflammatory Disorders: Is there a Case for Revis-iting Leukotrienes as Therapeutic Targets?, Inflammopharmacology, 14, 10 (2006); https://doi.org/10.1007/s10787-006-1496-6
  3. H.R. Chitme, M. Chandra and S. Kaushik, Studies on Anti-Diarrhoeal Activity of Calotropis Gigantea R.Br. in Experimental Animals, J. Pharm. Pharm. Sci., 7, 70 (2004).
  4. P.A. Akah and A.I. Nwambie, Evaluation of Nigerian Traditional Medicines: 1. Plants used for Rheumatic (Inflammatory) Disorders, J. Ethnopharmacol., 42, 179 (1994); https://doi.org/10.1016/0378-8741(94)90083-3
  5. R. Valsaraj, P. Pushpangadan, U.W. Smitt, A. Adsersen and U. Nyman, Antimicrobial Screening of Selected Medicinal Plants from India, J. Ethnopharmacol., 58, 75 (1997); https://doi.org/10.1016/S0378-8741(97)00085-8
  6. B. Mahesh and S. Sathish, Antimicrobial Activity of Some Important Medicinal Plant Against Plant and Human Pathogens, World J. Agric. Sci., 4, 839 (2008).
  7. M.K. Menon and A. Kar, Analgesic and Psychopharmacological Effects of the Gum Resin of Boswellia Serrata, Planta Med., 19, 333 (1971); https://doi.org/10.1055/s-0028-1099651
  8. M.Z. Siddiqui, Boswellia serrata, A Potential Antiinflammatory Agent: An Overview, Indian J. Pharm. Sci., 73, 255 (2011); https://doi.org/10.4103/0250-474X.93507
  9. K. Sengupta, J.N. Kolla, A.V. Krishnaraju, N. Yalamanchili, C.V. Rao, T. Golakoti, S. Raychaudhuri and S.P. Raychaudhuri, Cellular and Molecular Mechanisms of Anti-inflammatory Effect of Aflapin: A Novel Boswellia serrata Extract, Mol. Cell. Biochem., 354, 189 (2011); https://doi.org/10.1007/s11010-011-0818-1
  10. M.-T. Huang, V. Badmaev, Y. Ding, Y. Liu, J.-G. Xie and C.-T. Ho, Anti-Tumor and Anti-carcinogenic Activities of Triterpenoid, b-Boswellic Acid, Biofactors, 13, 225 (2000); https://doi.org/10.1002/biof.5520130135
  11. D. Poeckel and O. Werz, Boswellic Acids: Biological Actions and Molecular Targets, Curr. Med. Chem., 13, 3359 (2006); https://doi.org/10.2174/092986706779010333
  12. I.A. Siddiqui, F. Afaq, V.M. Adhami, N. Ahmad and H. Mukhtar, Antioxidants of the Beverage Tea in Promotion of Human Health, Antioxid. Redox Signal., 6, 571 (2004); https://doi.org/10.1089/152308604773934323
  13. C.L. Curtis, J.L. Harwood, C.M. Dent and B. Caterson, Biological Basis for the Benefit of Nutraceutical Supplementation in Arthritis, Drug Discov. Today, 9, 165 (2004); https://doi.org/10.1016/S1359-6446(03)02980-5
  14. L. Labban, Medicinal and Pharmacological Properties of Turmeric (Curcuma longa): A Review, Int. J. Pharm. Biomed. Sci., 5, 17 (2014).
  15. J. Cibere, Z. Deng, Y. Lin, R. Ou, Y. He, Z. Wang, A. Thome, A.J. Lehman, I.K. Tsang and J.M. Esdaile, A Randomized Double Blind, Placebo Controlled Trial of Topical Tripterygium Wilfordii in Rheumatoid Arthritis: Reanalysis Using Logistic Regression Analysis, J. Rheumatol., 30, 465 (2003).
  16. D. Qiu and P.N. Kao, Immunosuppressive and Anti-Inflammatory Mechanisms of Triptolide, The Principal Active Diterpenoid from the Chinese Medicinal Herb Tripterygium wilfordii Hook. f., Drugs R D, 4, 1 (2003); https://doi.org/10.2165/00126839-200304010-00001
  17. W.Z. Gu and S.R. Brandwein, Inhibition of Type II Collagen-Induced Arthritis in Rats by Triptolide, Int. J. Immunopharmacol., 20, 389 (1998); https://doi.org/10.1016/S0192-0561(98)00035-6
  18. B. Wang, L. Ma, X. Tao and P.E. Lipsky, Triptolide, An Active Component of the Chinese Herbal Remedy Tripterygium wilfordii Hook F, Inhibits Production of Nitric Oxide by Decreasing Inducible Nitric Oxide Synthase Gene Transcription, Arthritis Rheumatol., 50, 2995 (2004); https://doi.org/10.1002/art.20459
  19. J.M. Benni, R.N. Suresha and M.K. Jayanthi, Evaluation of the Anti-inflammatory Activity of Aegle marmelos (Bilwa) Root, Indian J. Pharmacol., 43, 393 (2011); https://doi.org/10.4103/0253-7613.83108
  20. J. Ojewole, Antinociceptive, Anti-inflammatory and Antidiabetic Effects of Bryophyllum pinnatum (Crassulaceae) Leaf Aqueous Extract, J. Ethnopharmacol., 99, 13 (2005); https://doi.org/10.1016/j.jep.2005.01.025
  21. R. Gupta, M. Lohani and S.K. Arora, Anti-inflammatory Activity of the Leaf Extracts/Fractions of Bryophyllum pinnatum Saliv. Syn, Int. J. Pharm. Sci. Rev. Res., 3, 16 (2010).
  22. A. Saha and M. Ahmed, The Analgesic and Anti-Inflammatory Activities of the Extract of Albizia aebbeck in Animal Model, Pak. J. Pharm. Sci., 22, 74 (2009).
  23. I. Raju, Mallika Moni and S. Venkataraman, Anti-Inflammatory and Antioxidant Activities of Cassia fistula Linn. Bark Extracts, Afr. J. Trad. Compl. Altern. Med., 2, 70 (2005); https://doi.org/10.4314/ajtcam.v2i1.31105
  24. G. Sreejith, P.G. Latha, V.J. Shine, G.I. Anuja, S.R. Suja, S. Sini, S. Shyma, S. Pradeep, P. Shika and S. Rajashekaran, Anti-Allergic, Anti-Inflammatory and Anti-Lipidperoxidant Effects of Cassia occidentalis Linn., Indian J. Exp. Biol., 48, 494 (2010).
  25. V.K. Garg and S.K. Paliwal, Anti-Inflammatory Activity of Aqueous Extract of Cynodon dactylon, Int. J. Pharmacol., 7, 370 (2011); https://doi.org/10.3923/ijp.2011.370.375
  26. M.Z. Asmawi, H. Kankaanranta, E. Moilanen and H. Vapaatalo, Anti-Inflammatory Activities of Emblica officinalis Gaertn Leaf Extracts, J. Pharm. Pharmacol., 45, 581 (1993); https://doi.org/10.1111/j.2042-7158.1993.tb05605.x
  27. V. Tomar, P. Kannojia, K.N. Jain and K.S. Dubey, Anti-Noceceptive and Anti-Inflammatory Activity of Leaves of Hibiscus - Rosa Sinensis, Int. J. Res. Ayurveda Pharm., 1, 201 (2010).
  28. K.S. Chandrashekar, A. Thakur and K.S. Prasanna, Anti-Inflammatory Activity of Moringa oleifera Stem Bark Extracts against Carrageenen Induced Rat Paw Edema, J. Chem. Pharm. Res., 2, 179 (2010).
  29. E.M. Franzotti, C.V. Santos, H.M. Rodrigues, R.H. Mourao, M.R. Andrade and A.R. Antoniolli, Anti-inflammatory, Analgesic Activity and Acute Toxicity of Sida cordifolia L. (Malva-branca), J. Ethnopharmacol., 72, 273 (2000); https://doi.org/10.1016/S0378-8741(00)00205-1
  30. R.L. Silva, G.B. Melo, V.A. Melo, Â.R. Antoniolli, P.R.T. Michellone, S. Zucoloto, M.A.N.C. Picinato, C.F.F. Franco, G.A. Mota and O. Castro e Silva, Effect of the Aqueous Extract of Sida cordifolia on Liver Regeneration after Partial Hepatectomy, Acta Cir. Bras., 21(suppl 1), 37 (2006); https://doi.org/10.1590/S0102-86502006000700009
  31. H. Shimoda, S.J. Shan, J. Tanaka, A. Seki, J.W. Seo, N. Kasajima, S. Tamura, Y. Ke and N. Murakami, Anti-Inflammatory Properties of Red Ginger (Zingiber officinale var. Rubra) Extract and Suppression of Nitric Oxide Production by Its Constituents, J. Med. Food, 13, 156 (2010); https://doi.org/10.1089/jmf.2009.1084
  32. L.K. Han, X.J. Gong, S. Kawano, M. Saito, Y. Kimura and H. Okuda, Anti-Obesity Effect of Ginger, Yakugaku Zasshi, 125, 213 (2005); https://doi.org/10.1248/yakushi.125.213
  33. G.A. Nogueira de Melo, R. Grespan, J.P. Fonseca, T.O. Farinha, E.L. da Silva, A.L. Romero, C.A. Bersani-Amado and R.K. Cuman, Inhibitory Effects of Ginger (Zingiber officinale Roscoe) Essential Oil on Leukocyte Migration in vivo and in vitro, J. Nat. Med., 65, 241 (2011); https://doi.org/10.1007/s11418-010-0479-5
  34. E.M. Anam, Anti-Inflammatory Activity of Compounds Isolated from the Aerial Parts of Abrus precatorius (Fabaceae), Phytomedicine, 8, 24 (2001); https://doi.org/10.1078/0944-7113-00001
  35. V.R. Mohan and K. Janardhanan, Chemical Determination of Nutritional and Antinutritional Properties in Tribal Pulses, J. Food Sci., 32, 465 (1995).
  36. R. Gautam, S.M. Jachak and A. Saklani, Anti-Inflammatory Effect of Ajuga bracteosa Wall Ex Benth. Mediated through Cyclooxygenase (COX) Inhibition, J. Ethnopharmacol., 133, 928 (2011); https://doi.org/10.1016/j.jep.2010.11.003
  37. W.K. Kayani, E. Dilshad, T. Ahmed, H. Ismail and B. Mirza, Evaluation of Ajuga bracteosa for Antioxidant, Anti-Inflammatory, Analgesic, Anti-depressant and Anticoagulant Activities, BMC Complement. Altern. Med., 16, 375 (2016); https://doi.org/10.1186/s12906-016-1363-y
  38. Y. Rao, S.H. Fang and Y.M. Tzeng, Antiinflammatory Activities of Flavonoids and a Triterpene Caffeate Isolated from Bauhinia variegata, Phytother. Res., 22, 957 (2008); https://doi.org/10.1002/ptr.2448
  39. R.N. Yadava and V.M. Reddy, Anti-Inflammatory activity of a Novel Flavonol Glycoside from the Bauhinia variegata Linn., Nat. Prod. Res., 7, 159 (2003); https://doi.org/10.1080/1478641031000104127
  40. S. Saha, E.V.S. Subrahmanyam, K.S. Chandrashekar and S.C. Shastry, in vivo Study for Anti-inflammatory Activity of Bauhinia variegata L. Leaves, Pharm. Crops, 2, 70 (2011); https://doi.org/10.2174/2210290601102010070
  41. M.A. Mohamed, M.R. Mammoud and H. Hayen, Evaluation of Anti-nociceptive and Anti-inflammatory Activities of a New Triterpene Saponin from Bauhinia variegata Leaves, Z Naturforsch C J Biosci., 64, 798 (2009); https://doi.org/10.1515/znc-2009-11-1208
  42. S.M. Bairagi, A.A. Aher, N. Nema and P.K. Nimase, Anti-Inflammatory Evaluation of Methanol Extract and Aqueous Fraction of the Bark of Bauhinia variegata (Leguminosae), Int. J. Res. Pharm. Chem., 2, 77 (2012).
  43. C.H. Lu, Y.Y. Li, L.J. Li, L.Y. Liang and Y.M. Shen, Anti-Inflammatory Activities of Fractions from Geranium nepalense and Related Polyphenols, Drug Discov Ther., 6, 194 (2012).
  44. J.A. Ojewole, Antinociceptive, Anti-Inflammatory and Antidiabetic Effects of Leonotis leonurus (L.) R. Br. [Lamiaceae] Leaf Aqueous Extract in Mice and Rats, Methods Find. Exp. Clin. Pharmacol., 27, 257 (2005); https://doi.org/10.1358/mf.2005.27.4.893583
  45. M.A. El-Ansari, E.S.A. Aboutabl, A.R.H. Farrag, M. Sharaf, U.W. Hawas, G.M. Soliman and G.S. El-Seed, Phytochemical and Pharma-cological Studies on Leonotis leonurus, Pharm. Biol., 47, 894 (2009); https://doi.org/10.1080/13880200902942428
  46. Y. Frum and A.M. Viljoen, in vitro 5-Lipoxygenase and Anti-Oxidant Activities of South African Medicinal Plants Commonly Used Topically for Skin Diseases, Skin Pharmacol. Physiol., 19, 329 (2006); https://doi.org/10.1159/000095253
  47. A. Diantini, A. Subarnas, K. Lestari, E. Halimah, Y. Susilawati, E. Supriyatna, E. Julaeha, T.H. Achmad, E.W. Suradji, C. Yamazaki, K. Kobayashi, H. Koyama and R. Abdulah, Kaempferol-3-O-rhamnoside Isolated from the Leaves of Schima wallichii Korth. Inhibits MCF-7 Breast Cancer Cell Proliferation through Activation of the Caspase Cascade Pathway, Oncol. Lett., 3, 1069 (2012); https://doi.org/10.3892/ol.2012.596
  48. S. Dewanjee, V. Mandal, R. Sahu, T.K. Dua, A. Manna and S.C. Mandal, Anti-inflammatory Activity of a Polyphenolic Enriched Extract of Schima wallichii Bark, Nat. Prod. Res., 25, 696 (2011); https://doi.org/10.1080/14786410802560732
  49. M.I. Barliana, E.W. Suradji, R. Abdulah, A. Diantini, T. Hatabu, J.N. Shimada, A. Subarnas and H. Koyama, Antiplasmodial Properties of Kaempferol-3-O-rhamnoside Isolated from the Leaves of Schima wallichii against Chloroquine-Resistant Plasmodium falciparum, Biomed. Rep., 2, 579 (2014); https://doi.org/10.3892/br.2014.271
  50. C.A. Williams, F. Goldstone and J. Greenham, Flavonoids, Cinnamic Acids and Coumarins from the Different Tissues and Medicinal Prepar-ations of Taraxacum officinale, Phytochemistry, 42, 121 (1996); https://doi.org/10.1016/0031-9422(95)00865-9
  51. M. Hfaiedh, D. Brahmi and L. Zourgui, Hepatoprotective Effect of Taraxacum officinale Leaf Extract on Sodium Dichromate-Induced Liver Injury in Rats, Environ Toxicol., 31, 339 (2016); https://doi.org/10.1002/tox.22048
  52. B.A. Clare, R.S. Conroy and K. Spelman, The Diuretic Effect in Human Subjects of an Extract of Taraxacum officinale Folium over a Single Day, J. Altern. Complement. Med., 15, 929 (2009); https://doi.org/10.1089/acm.2008.0152
  53. Y.J. Koh, D.S. Cha, J.S. Ko, H.J. Park and H.D. Choi, Anti-inflammatory Effect of Taraxacum officinale Leaves on Lipopolysaccharide-Induced Inflammatory Responses in RAW 264.7 Cells, J. Med. Food, 13, 870 (2010); https://doi.org/10.1089/jmf.2009.1249
  54. W.R. Jr Henderson, The Role of Leukotrienes in Inflammation, Ann. Intern. Med., 121, 684 (1994); https://doi.org/10.7326/0003-4819-121-9-199411010-00010
  55. J.C. Adkins and R.N. Brogden, A Review of its Pharmacology and Thera-peutic Potential in the Management of Asthma, Drugs, 55, 121 (1998); https://doi.org/10.2165/00003495-199855010-00008
  56. P.N. Dekhuijzen and P.P. Koopmans, Pharmacokinetic Profile of Zafirlukast, Clin Pharmacokinet., 41, 105 (2002); https://doi.org/10.2165/00003088-200241020-00003
  57. N.T. Chandrika, M.Y. Fosso, Y. Alimova, A. May, O.A. Gonzalez and S. Garneau-Tsodikova, Novel Zafirlukast Derivatives Exhibit Selective Antibacterial Activity against Porphyromonas gingivalis, MedChemComm, 10, 926 (2019); https://doi.org/10.1039/C9MD00074G
  58. P.E. Malo, R.L. Bell, T.K. Shaughnessy, J.B. Summers, D.W. Brooks and G.W. Carter, The 5-Lipoxygenase Inhibitory Activity of Zileuton in in vitro and in vivo Models of Antigen-Induced Airway Anaphylaxis, Pulm Pharmacol., 7, 73 (1994); https://doi.org/10.1006/pulp.1994.1008
  59. C.C. Zouboulis, Zileuton, A New Efficient and Safe Systemic Anti-acne Drug, Dermatoendocrinol., 1, 188 (2009); https://doi.org/10.4161/derm.1.3.8368
  60. R. Anderson, A.J. Theron, C.M. Gravett, H.C. Steel, G.R. Tintinger and C. Feldman, Montelukast Inhibits Neutrophil Pro-inflammatory Activity by a Cyclic AMP-Dependent Mechanism, Br. J. Pharmacol., 156, 105 (2009); https://doi.org/10.1111/j.1476-5381.2008.00012.x
  61. S.J. Keam, K.A. Lyseng-Williamson and K.L. Goa, A Review of its Use in the Management of Asthma, Drugs, 63, 991 (2003); https://doi.org/10.2165/00003495-200363100-00005
  62. S. Yoshida, Y. Ishizaki, T. Shoji, K. Onuma, H. Nakagawa, M. Nakabayashi, K. Akahori, H. Hasegawa and H. Amayasu, Effect of Pranlukast on Bronchial Inflammation in Patients with Asthma, Clin. Exp. Allergy, 30, 1008 (2000); https://doi.org/10.1046/j.1365-2222.2000.00834.x
  63. V. Capra, M. Bolla, P.A. Belloni, M. Mezzetti, G.C. Folco, S. Nicosia and G.E. Rovati Pharmacological Characterization of the Cysteinyl-Leukotriene Antagonists CGP 45715A (Iralukast) and CGP 57698 in Human Airways in vitro, Br. J. Clin. Pharmacol., 123, 590 (1998); https://doi.org/10.1038/sj.bjp.0701636
  64. S.R. O’Donnell, Leukotrienes - Biosynthesis and Mechanisms of Action, Aust. Prescr., 22, 55 (1999); https://doi.org/10.18773/austprescr.1999.053
  65. E. Adelroth, M.D. Inman, E. Summers, D. Pace, M. Modi and P.M. O’Byrne, Prolonged Protection Against Exercise-Induced Bronchocon-striction by the Leukotriene D 4–Receptor Antagonist Cinalukast, J. Allergy Clin. Immunol., 99, 210 (1997); https://doi.org/10.1016/S0091-6749(97)70098-8
  66. J.-W.J. Lammers, P. Van Daele, F.M.J. Van den Elshout, M. Decramer, A. Buntinx, I. De Lepeleire and B. Friedman, Bronchodilator Properties of an Inhaled Leukotriene D4 Antagonist (Verlukast-MK-0679) in Asthmatic Patients, Pulm. Pharmacol., 5, 121 (1992); https://doi.org/10.1016/0952-0600(92)90029-G
  67. K.F. Chung, Leukotriene Receptor Antagonists and Biosynthesis Inhibitors: Potential Breakthrough in Asthma Therapy, Eur. Respir. J., 8, 1203 (1995); https://doi.org/10.1183/09031936.95.08071203
  68. H.E. Claesson and S.E. Dahlen, Asthma and Leukotrienes: Anti-leukotrienes as Novel Anti-asthmatic Drugs, J. Intern. Med., 245, 205 (1999); https://doi.org/10.1046/j.1365-2796.1999.00418.x
  69. N. Barnes, Leukotriene Receptor Antagonists: Clinical Effects, J. R. Soc. Med., 90, 200 (1997); https://doi.org/10.1177/014107689709000405