H2S Donors in creation of innovative drugs

Authors

  • D. V. Kaminskyy Lviv national medical University Daniel Galitsky, Ukraine
  • A. P. Kryshchyshyn Lviv national medical University Daniel Galitsky, Ukraine
  • O. P. Yelisyeyeva Lviv national medical University Daniel Galitsky, Ukraine
  • R. B. Lesyk Lviv national medical University Daniel Galitsky, Ukraine

DOI:

https://doi.org/10.24959/ophcj.17.918

Keywords:

H2S, metabolism, biological effects of H2S, sulfur-containing groups, H2S-donors

Abstract

Fundamental studies have identified a new group of gaseous signaling molecules – the so-called gasotransmitters – NO, CO, and H2S, which are involved in the regulation of a large number of metabolic processes. The results of these studies allowed determining a new direction in medicinal/pharmaceutical chemistry – creation of hydrogen sulfide donor compounds as potential drugs. The article presents the main achievements in the search for new H2S donors: the main stages of H2S metabolism and its biological effects; the classes of compounds that can release hydrogen sulfide based on the nature of sulfur-containing functional groups as well as the mechanism of H2S releasing. Additionally, the characteristic of the most successful direction – creation of the so-called hybrid molecules is given. The latter are compounds bearing fragments of the well known drugs covalently bounded with groups being capable to release H2S.

Downloads

Download data is not yet available.

References

  1. Lowicka, E. Hydrogen sulfide (H(2)S) – the third gas of interest for pharmacologists / E. Lowicka, J. Beltowski // Pharmacol. Reports. – 2007. – Vol. 59, Issue 1. – P. 4–24. Available at : http://if pan.krakow.pl/pjp/pdf/2007/1_4.pdf
  2. Li, L. Actions and interactions of nitric oxide, carbon monoxide and hydrogen sulphide in the cardiovascular system and in inflammation – a tale of three gases! / L. Li, A. Hsu, P. K. Moore // Pharmacol. & Therapeutics. – 2009. – Vol. 123, Issue 3. – P. 386–400. doi: 10.1016/j.pharmthera.2009.05.005
  3. Pryor, W. A. Free radical biology and medicine : it’s a gas, man! / W. A. Pryor // AJP : Regulatory, Integrative and Comparative Physiol. – 2006. – Vol. 291, Issue 3. – P. R491–R511. doi: 10.1152/ajpregu.00614.2005
  4. Abe, K. The possible role of hydrogen sulfide as an endogenous neuromodulator / K. Abe, H. J. Kimura // Neurosci. – 1996. – Vol. 16, Issue 3. – P. 1066–1071. Available at : http://www.jneurosci.org/content/jneuro/16/3/1066.full.pdf
  5. Brosnan, J. T.The sulfur–containing amino acids : an overview / J. T. Brosnan, M. E. Brosnan // J. Nutrit. – 2006. – Vol. 136, Issue 6. – P. 1636–1640. Available at : http://jn.nutrition.org/content/136/6/1636S.full.pdf+html
  6. Impaired homocysteine metabolism and atherothrombotic disease / P. Durand, M. Prost, N. Loreau et al. // D. Lab. Invest. – 2001. – Vol. 81, Issue 5. – P. 645–672. Available at : http://www.nature.com/labinvest/journal/v81/n5/pdf/3780275a.pdf
  7. Hayes, K. C. Taurine in metabolism / K. C. Hayer, J. A. Sturman // Ann. Rev. Nutr. – 1981. – Vol. 1. – P. 401–425.
  8. Kabil, O. Enzymology of H2S Biogenesis, Decay and Signaling / O. Kabil, R. Banerjee // Antiox. Redox. Signal. – 2014. – Vol. 20, Issue 5. – P. 770–782. doi: 10.1089/ars.2013.5339
  9. Kimura, H. Hydrogen sulfide : its production and functions / H. Kimura // Exp. Physiol. – 2011. – Vol. 96, Issue 9. – P. 833–835. doi: 10.1113/expphysiol.2011.057455
  10. Kimura, H. Physiological role of hydrogen sulfide and polysulfide in the central nervous system / H. Kimura // Neurochem. Int. – 2013. – Vol. 63,
  11. Issue 5. – P.492–497. doi: 10.1016/j.neuint.2013.09.003
  12. Dynamic Change of Hydrogen Sulfide After Traumatic Brain Injury and its Effect in Mice / M. Zhang, H. Shan, T. Wang et al. // Neurochem. Res. – 2013. – Vol. 38, Issue 4. – P.714–725.doi: 10.1007/s11064–013–0969–4
  13. Vascular Endothelium Expresses 3–Mercaptopyruvate Sulfurtransferase and Produces Hydrogen Sulfide / N. Shibuya, Y. Mikami, Y. Kimura et al. // J. Biochem. – 2009. – 146, Issue 5. – P. 623–626. doi: 10.1093/jb/mvp111
  14. Wang, R. Physiological Implications of Hydrogen Sulfide : A Whiff Exploration That Blossomed / R. Wang // Physiol. Rev. – 2012. – Vol. 92, Issue 2. – P.791–896. doi: 10.1152/physrev.00017.2011
  15. A novel pathway for the production of hydrogen sulfide from D–cysteine in mammalian cells / N. Shibuya, S. Koike, M. Tanaka et al. // Nature Commun. – 2013. – Vol. 4. – 1366 p. doi: 10.1038/ncomms2371
  16. Jakubowski, H. Synthesis of homocysteine thiolactone by methionyl–tRNA synthetase in cultured mammalian cells / H. Jakubowski, E. Goldman // FEBS Lett. – 1993. – Vol. 317, Issue 3. – P. 237–240. doi: 10.1016/0014–5793(93)81283–6
  17. Eto, K. The production of hydrogen sulfide is regulated by testosterone and S–adenosyl–l–methionine in mouse brain / K. Eto, H. Kimura // J. Neurochem. –2002. – Vol. 83, Issue 1. – P.80–86. doi: 10.1046/j.1471–4159.2002.01097.x
  18. Obeid, R. Mechanisms of homocysteine neurotoxicity in neurodegenerative diseases with special reference to dementia / R. Obeid, W. Herrmann // FEBS Lett. – 2006. – Vol. 580, Issue 13. – P. 2994–3005. doi: 10.1016/j.febslet.2006.04.088
  19. Kimura, H. Hydrogen Sulfide Is a Signaling Molecule and a Cytoprotectant / H. Kimura, Y. Kimura, N. Shibuya // Antioxid. Redox. Signal. – 2012. – Vol. 17, Issue 1. – P.45–57. doi: 10.1089/ars.2011.4345
  20. Thioredoxin and dihydrolipoic acid are required for 3–mercaptopyruvate sulfurtransferase to produce hydrogen sulfide / Y. Mikami, N. Shibuya, Y. Kimura et al. // Biochem. J. – 2011. – Vol. 439, Issue 3. – P. 479–485. doi: 10.1042/BJ20110841
  21. Qian, L. Chemical foundations of hydrogen sulfide biology / L. Qian, J. R. Lancaster // Nitric Oxide. – 2013. – Vol. 35. – P. 21–34. doi: 10.1016/j. niox.2013.07.001
  22. Zhao, Y. Hydrogen sulfide (H2S) releasing agents : chemistry and biological applications / Y. Zhao, T. D. Biggs, M. Xian // Chem. Comm. – 2014. – Vol. 50, Issue 8. – P. 11788–11805. doi: 10.1039/C4CC00968A
  23. Natural Products Containing Hydrogen Sulfide Releasing Moieties / M. D. Pluth, T. S. Bailey, M. D. Hammers et al. // Synlett. – 2015. – Vol. 26, Issue 19. – P. 2633–2643. doi: 10.1055/s–0035–1560638
  24. Polysulfides are possible H2S–derived signaling molecules in rat brain / Y. Kimura, Y. Mikami, K. Osumi et al. // FASEB J. – 2013. – Vol. 27, Issue 6. – P. 2451–2457. doi: 10.1096/fj.12–226415
  25. Kimura, H. Hydrogen sulfide : its production, release and functions / H. Kimura // Amino Acids. – 2011. – Vol. 41, Issue 1. – P. 113–121. doi: 10.1007/s00726–010–0510–x
  26. Hughes, M. N. Making and working with hydrogen sulfide / M. N. Hughes, M. N. Centelles, K. P. Moore // Free Radic. Biol. Med. – 2009. – Vol. 47, Issue 10. – P. 1346–1353. doi: 10.1016/j.freeradbiomed.2009.09.018
  27. Guo, W. Hydrogen sulfide and translational medicine / W. Guo, Z. Y. Cheng, Y. Z. Zhu // Acta Pharmacol. Sinica. – 2013. – Vol. 34, Issue 10. – P. 1284–1291. doi: 10.1038/aps.2013.127
  28. Kashfi, K. Biology and therapeutic potential of hydrogen sulfide and hydrogen sulfide–releasing chimeras / K. Kashfi, K. R. Olson // Biochem. Pharmacol. 2013. – Vol. 85, Issue 5. – P. 689–703. doi: 10.1016/j.bcp.2012.10.019
  29. Reactivity of hydrogen sulfide with peroxynitrite and other oxidants of biological interest / M. Carballal, E. Trujillo, S. Cuevasanta et al. // Free Rad. Biol. Med. – 2011. – Vol. 50, Issue 1, P. 196–205. doi: 10.1016/j.freeradbiomed.2010.10.705
  30. Zhao, Y. Cysteine–Activated Hydrogen Sulfide (H2S) Donors / Y. Zhao, H. Wang, M. J. Xian // Am. Chem. Soc. – 2011. – Vol. 133, Issue 1. – P. 15–17. doi: 10.1021/ja1085723
  31. Wang, R. Two’s company, three’s a crowd : can H2S be the third endogenous gaseous transmitter? / R. Wang // FASEB J. – 2002. – Vol. 16, Issue 13. – P. 1792–1798. doi: 10.1096/fj.02–0211hyp
  32. The vasorelaxant effect of H2S as a novel endogenous gaseous KATP channel opener / W. Zhao, J. Zhang, Y. Lu, R. Wang // EMBO J. – 2001. – Vol. 20, Issue 21. – P. 6008–6016. doi: 10.1093/emboj/20.21.6008
  33. Tang, G. Direct stimulation of KATP channels by exogenous and endogenous hydrogen sulfide in vascular smooth muscle / G. Tang // Mol. Pharmacol. –2005. – Vol. 68. – P. 1757–1764. doi: 10.1124/mol.105.017467
  34. Zhao, W. H2S–induced vasorelaxation and underlying cellular and molecular mechanisms / W. Zhao, R. Wang // Am. J. Physiol. Heart Circ. Physiol. – 2002. – Vol. 283, Issue 2. – P. H474–H480. doi: 10.1152/ajpheart.00013.2002
  35. Hydrogen sulfide in combination with taurine or cysteic acid reversibly abolishes sodium currents in neuroblastoma cells / M. W. Warenycia, J. A. Steele, E. Karpinski, R. J. Reiffenstein // Neurotox. – 1989. – Vol. 10. – P. 191–199.
  36. Biomarkers of oxidative and nitro–oxidative stress: conventional and novel approaches / A. Cipak Gasparovic, N. Zarkovic, K. Zarkovic et al. // Brit. J. Pharmacol. – 2017. doi: 10.1111/bph.13673
  37. Vasoconstrictive effect of hydrogen sulfide involves downregulation of cAMP in vascular smooth muscle cells / J. J. Lim, Y.–H. Liu et al. // Am. J. Physiol. Cell Physiol. – 2008. – Vol. 295, Issue 5. – P. C1261–C1270. doi: 10.1152/ajpcell.00195.2008
  38. Hydrogen Sulfide–Induced Dual Vascular Effect Involves Arachidonic Acid Cascade in Rat Mesenteric Arterial Bed / R. d’E. Di Villa Bianca, R. Sorrentino, C. Coletta et al. // J. Pharmacol. Exp. Ther. – 2011. –Vol. 337, Issue 1. – P. 59–64. doi: 10.1124/jpet.110.176016
  39. The Role of Endogenous H2S in Cardiovascular Physiology / N. Skovgaard, A. Gouliaev, M. Aalling, U. Simonsen // Curr. Pharm. Biotechnol. – 2011. – Vol. 12, Issue 9. – P. 1385–1393. doi: 10.2174/138920111798280956
  40. Contractile and Vasorelaxant Effects of Hydrogen Sulfide and Its Biosynthesis in the Human Internal Mammary Artery / G. D. Webb, L. H. Lim, V. M. S. Oh et al. // J. Pharmacol. Exp. Ther. – 2008. – Vol. 324, Issue 2. – P. 876–882. doi: 10.1124/jpet.107.133538
  41. Lavu, M. Hydrogen sulfide–mediated cardioprotection : mechanisms and therapeutic potential / M. Lavu, S. Bhushan, D. J. Lefler // Clin. Sci. – 2011. – Vol. 120, Issue 6. – P. 219–229. doi: 10.1042/CS20100462
  42. Brittain, T. The interaction of human neuroglobin with hydrogen sulphide / T. Brittain, Y. Yosaatmadja, K. Henty // IUBMB Life. – 2008. – Vol. 60, Issue 2. – P. 135–138.doi: 10.1002/iub.16
  43. Hydrogen sulfide mitigates matrix metalloproteinase–9 activity and neurovascular permeability in hyperhomocysteinemic mice / N. Tyagi, S. Givvimani, N. Qipshidze et al. // Neurochem. Int. – 2010. – Vol. 56, Issue 2. – P. 301–307. doi: 10.1016/j.neuint.2009.11.002
  44. Calvert, J. W. Novel Insights Into Hydrogen Sulfide–Mediated Cytoprotection / J. W. Calvert, W. A. Coetzee, D. J. Lefer //Antioxid. Redox. Signal. – 2010. – Vol. 12, Issue 10. – P. 1203–1217. doi: 10.1089/ars.2009.2882
  45. Physiological and pharmacological features of the novel gasotransmitter : Hydrogen sulfide / D. Mancardi, C. Penna, A. Merlino et al. // Biochim. Biophys. Acta. – 2009. – Vol. 1787, Issue 7. – P. 864–872. doi: 10.1016/j.bbabio.2009.03.005
  46. Whiteman, M. Hydrogen sulfide and the vasculature: a novel vasculoprotective entity and regulator of nitric oxide bioavailability? / M. Whiteman, P. K. Moore // J. Cell Mol. Med. – 2009. – Vol. 13, Issue 3. – P. 488–507. doi: 10.1111/j.1582–4934.2009.00645.x
  47. Chemical probes for molecular imaging and detection of hydrogen sulfide and reactive sulfur species in biological systems / V. S. Lin, W. Chen, M. Xian, C. J. Chang // Chemical. Soc. Rev. – 2015. – Vol. 44, Issue 14. – P. 4596–4618. doi: 10.1039/C4CS00298A
  48. Юрченко, П. О. Роль системи гідроген сульфіду в механізмах ураження мозку за умов гіпергомоцистеїнемії : дис. … н. ст. канд. мед. наук : 14.01.32 / П. О. Юрченко. – Вінниця, 2016.
  49. Пентюк, Н. О. Гіперпродукція вазоактивних медіаторів як патогенетичний чинник розвитку ускладнень цирозу печінки у щурів / Н. О. Пентюк, Н. В. Харченко // Сучасна гастроентерол. – 2010. – № 2 (52), С. 33–43. Режим доступу : http://vitapol.com.ua/user_files/pdfs/gastro/978639636524872_06062010115352.pdf
  50. Li, L. Could hydrogen sulfide be the next blockbuster treatment for inflammatory disease? / L. Li, P. K. Moore // Expert. Rev. Clin. Pharmacol. – 2013. – Vol. 6, Issue 6. – P. 593–595. doi: 10.1586/17512433.2013.842126
  51. Wallace, J. L. Hydrogen sulfide–releasing anti–inflammatory drugs / J. L. Wallace // Trends Pharmacol. Sci. – 2007. – Vol. 28, Issue 10. – P. 501–505. doi: 10.1016/j.tips.2007.09.003
  52. Hydrogen sulfide induces ICAM–1 expression and neutrophil adhesion to caerulein–treated pancreatic acinar cells through NF–κB and Src–family kinases pathway / R. Tamizhselvi, Y. H. Koh, J. Sun et al. // Exp. Cell Res. – 2010. – Vol. 316, Issue 9. – P. 1625–1636. doi: 10.1016/j.yexcr.2010.02.044
  53. Pro–inflammatory effects of hydrogen sulphide on substance P in caerulein–induced acute pancreatitis / M. Bhatia, J. N. Sidhapuriwala et al. // J. Cell Mol. Med. – 2008. – Vol. 12, Issue 2. – P. 580–590. doi: 10.1111/j.1582–4934.2007.00131.x
  54. Hydrogen sulfide donors in research and drug development / Z. J. Song, M. Y. Ng, Z. W. Lee et al. // Med. Chem. Comm. – 2014. – Vol. 5, Issue 5. – 557 p. doi: 10.1039/C3MD00362K
  55. Forgan, L. G. Oxygen consumption, ventilation frequency and cytochrome c oxidase activity in blue cod (Parapercis colias) exposed to hydrogen sulphide or isoeugenol / L. G. Forgan, M. E. Forster // Comp. Biochem. Physiol. Toxicol. Pharmacol : CBP. – 2010. – Vol. 151, Issue 1. – P. 57–65. doi: 10.1016/j.cbpc.2009.08.008
  56. Baumgart, K. Applying gases for microcirculatory and cellular oxygenation in sepsis : effects of nitric oxide, carbon monoxide, and hydrogen sulfide / K. Baumgart, P. Radermacher, F. Wagner // Curr. Opin. Anaesthesiol. – 2009. – Vol. 22, Issue 2. – P. 168–176. doi: 10.1097/ACO.0b013e328328d22f
  57. H2S during circulatory shock : Some unresolved questions / O. McCook, P. Radermacher, C. Volani et al. // Nitric Oxide. – 2014. – Vol. 41, P. 48–61. doi: 10.1016/j.niox.2014.03.163
  58. Hartle, M. D. A practical guide to working with H2S at the interface of chemistry and biology / M. D. Hartle, M. D. Pluth // Chem. Soc. Rev. – 2016. – Vol. 45, Issue 2. – P. 6108–6117. doi: 10.1039/C6CS00212A
  59. Wallace, J. L. Hydrogen sulfide–based therapeutics: exploiting a unique but ubiquitous gasotransmitter / J. L. Wallace, R. Wang // Nat. Rev. Drug Discov. –2015. – Vol. 14, Issue 5. – P. 329–345. doi: 10.1038/nrd4433
  60. Onions? A global benefit to health / G. Griffiths, L. Trueman et al. // Phytother. Res. – 2002. – Vol. 16, Issue 7. – P. 603–615. doi: 10.1002/ptr.1222
  61. Смаглий, Л. Сероводород – новое лекарство для сосудов / Л. Смаглий // Биомолекула. – 2013. Режим доступа : http://biomolecula.ru/content/1373.
  62. Synthesis and Biological Effects of Hydrogen Sulfide (H2S) : Development of H2S–Releasing Drugs as Pharmaceuticals / G. Caliendo, G. Cirino, V. Santagada, J. L. Wallace // J. Med. Chem. – 2010. – Vol. 53, Issue 17. – P. 6275–6286. doi: 10.1021/jm901638j
  63. Phosphinodithioate and Phosphoramidodithioate Hydrogen Sulfide Donors / M. Whiteman, A. Perry, Z. Zhou et al. // Handbook of Exp. Pharmacol. – 2015. – Vol. 230. – P. 337–363. doi: 10.1007/978–3–319–18144–8_17
  64. Characterization of a Novel, Water–Soluble Hydrogen Sulfide–Releasing Molecule (GYY4137) : New Insights Into the Biology of Hydrogen Sulfide / L. Li, M. Whiteman, Y. Y. Guan et al. // Circulation. – 2008. – Vol. 117, Issue 18. – P. 2351–2360. doi: 10.1161/CIRCULATIONAHA.107.753467
  65. The Slow–Releasing Hydrogen Sulfide Donor, GYY4137, Exhibits Novel Anti–Cancer Effects In Vitro and In Vivo / Z. W. Lee, J. Zhou et al. // PLoS One. – 2011. – Vol. 6, Issue 6. – e21077 p. doi: 10.1371/journal.pone.0021077
  66. Synthesis and evaluation of phosphorodithioate–based hydrogen sulfide donors / C. Park, Y. Zhao, Z. Zhu et al. // Mol. Bio. Syst. – 2013. – Vol. 9, Issue 10. –P. 2430–2434. doi: 10.1039/C3MB70145J
  67. Kodela, R. NOSH–Aspirin : A Novel Nitric Oxide–Hydrogen Sulfide–Releasing Hybrid: A New Class of Anti–inflammatory Pharmaceuticals / R. Kodela, M. Chattopadhyay, K. Khosrow // ACS Med. Chem. Lett. – 2012. – Vol. 3, Issue 3. – P. 257–262. doi: 10.1021/ml300002m
  68. Enhanced chemopreventive effects of a hydrogen sulfide–releasing anti–inflammatory drug (ATB–346) in experimental colorectal cancer / W. Elsheikh, R. W. Blackler, K. L. Flannigan, J. L. Wallace // Nitric Oxide. – 2014. – Vol. 41. – P.131–137. doi: 10.1016/j.niox.2014.04.006
  69. Thioglycine and l–thiovaline : Biologically active H2S–donors / Z. Zhou, M. von Wantoch Rekowski, C. Coletta, C. Szabo et al. // Bioorg. Med. Chem. – 2012. – Vol. 20, Issue 8. – P. 2675–2678. doi: 10.1016/j.bmc.2012.02.028
  70. Light–Induced Hydrogen Sulfide Release from “Caged” gem–Dithiols / N. O. Devarie–Baez, P. E. Bagdon, B. Peng et al. // Org. Lett. – 2013. – Vol. 15, Issue 11. – P. 2786–2789. doi: 10.1021/ol401118k
  71. New Biologically Active Hydrogen Sulfide Donors / T. Roger, F. Raynaud et al. // ChemBioChem. – 2013. – Vol. 14, Issue 17. – P. 2268–2271. doi: 10.1002/cbic.201300552
  72. Synthesis of a photocontrollable hydrogen sulfide donor using ketoprofenate photocages / N. Fukushima, N. Ieda et al. // Chem. Commun. – 2014. – Vol. 50, Issue 5. – P. 587–589. doi: 10.1039/C3CC47421F
  73. Anti–inflammatory and gastrointestinal effects of a novel diclofenac derivative / L. Li, G. Rossoni et al. // Free Rad. Biol. Med. – 2007. – Vol. 42, Issue 5. – P. 706–719. doi: 10.1016/j.freeradbiomed.2006.12.011
  74. Hydrogen Sulfide Releasing Aspirin, ACS14, Attenuates High Glucose–Induced Increased Methylglyoxal and Oxidative Stress in Cultured Vascular Smooth Muscle Cells / Q. Huang, A. Sparatore, P. Del Soldato et al. // PloS ONE. – 2014. – Vol. 9, Issue 6. – e97315 p. doi: 10.1371/journal.pone.0097315
  75. NOSH–aspirin (NBS–1120), a novel nitric oxide– and hydrogen sulfide–releasing hybrid is a potent inhibitor of colon cancer cell growth in vitro and in a xenograft mouse model / M. Chattopadhyay, R. Kodela, K. R., Olson, K. Kashfi // Biochem. Biophys. Res. Commun. – 2012. – Vol. 419, Issue 3. – P. 523–528. doi: 10.1016/j.bbrc.2012.02.051
  76. Elsey, D. J. Regulation of cardiovascular cell function by hydrogen sulfide (H2S) / D. J. Elsey, R. C. Fowkes, G. F. Baxter // Cell Biochem. Func. – 2010. – Vol. 28, Issue 2. – P. 95–106. doi: 10.1002/cbf.1618
  77. Hydrogen sulfide attenuates neurodegeneration and neurovascular dysfunction induced by intracerebral–administered homocysteine in mice / P. K. Kamat, A. Kalani, S. Givvimani et al. // Neurosci. – 2013. – Vol. 252. – P. 302–319. doi: 10.1016/j.neuroscience.2013.07.051
  78. Aruoma, O. I. Protection Against Oxidative Damage and Cell Death by the Natural Antioxidant Ergothioneine / O. I. Aruoma, J. P. E. Spencer, N. Mahmood // Food Chem. Toxicol. – 1999. – Vol. 37, Issue 11. – P. 1043–1053. doi: 10.1016/S0278–6915(99)00098–8
  79. Mitsuyama, H. Uptake and antioxidant effects of ergothioneine in human erythrocytes / H. Mitsuyama, J. M. May // Clin. Sci. – 1999. – Vol. 97, Issue 4. – P. 407–411. doi: 10.1042/cs0970407
  80. Lesyk, R. B. 4–Thiazolidones : Centenarian History, Current Status and Perspectives for Modern Organic and Medicinal Chemistry / R. B. Lesyk, B. S. Zimenkovsky // Curr. Org. Chem. – 2004. – Vol. 8, Issue 16. – P. 1547–1577. doi: 10.2174/1385272043369773
  81. Пат. України на корисну модель G 09 B 23/28. Спосіб зниження ульцерогенної дії нестероїдних протизапальних препаратів на експериментальних моделях у щурів / Ільків, І. І., Лесик, Р. Б., Скляров, О. Я. – 108412 ; опубл. 11.07.2016, Бюл. № 13.
  82. Ilkiv, I. Evaluation of novel 4–thiazolidinone–based derivatives as possible cytoprotective agents against stress model in rats / I. Ilkiv, R. Lesyk, O. J. Sklyarov // Appl. Pharm. Sci. – 2017. – Vol. 7, Issue 01. – P. 199–203. doi: 10.7324/JAPS.2017.70129

Published

2017-05-23

How to Cite

(1)
Kaminskyy, D. V.; Kryshchyshyn, A. P.; Yelisyeyeva, O. P.; Lesyk, R. B. H2S Donors in Creation of Innovative Drugs. J. Org. Pharm. Chem. 2017, 15, 27-46.

Issue

Section

Original Researches