Selective Carboxylesterase Inhibitors for Improving Efficacy, Safety and Rational use of Ester-Containing Drugs

Main Article Content

N.P. Boltneva
G.F. Makhaeva
E.V. Shchegol’kov
Ya.V. Burgart
V.I. Saloutin

Abstract

In clinical practice, a large number of prodrugs and active drugs containing an ester, carbamate or amide moiety are used. Carboxylesterase (CaE, EC 3.1.1.1) is the key enzyme of hydrolytic metabolism of such drugs in the body, it largely determines their pharmacokinetics, bioavailability, efficacy and possible toxic effects. Using CaE selective inhibitors as components of combined drug therapy it is possible us to regulate the rate of hydrolytic transformation of ester-containing drugs and opens the possibility of their rational use. The development of effective and selective CaE inhibitors suitable for in vivo application is a new promising approach in medicinal chemistry and pharmacology that allows to improve the efficacy, bioavailability and reduce the side effects of ester-containing drugs.

Article Details

How to Cite
Boltneva, N., Makhaeva, G., Shchegol’kov, E., Burgart, Y., & Saloutin, V. (2018). Selective Carboxylesterase Inhibitors for Improving Efficacy, Safety and Rational use of Ester-Containing Drugs. Biomedical Chemistry: Research and Methods, 1(3), e00026. https://doi.org/10.18097/BMCRM00026
Section
REVIEWS

References

  1. Laizure, S.C., Herring, V., Hu, Z., Witbrodt, K., & Parker, R.B. (2013) The role of human carboxylesterases in drug metabolism: have we overlooked their importance? Pharmacotherapy, 33(2), 210-222. DOI
  2. Neuvonen, M., Tarkiainen, E.K., Tornio, A., Hirvensalo, P., Tapaninen, T., Paile-Hyvarinen, M., Itkonen, M. K., Holmberg, M.T., Karja, V., Mannisto, V.T., Neuvonen, P.J., Pihlajamaki, J., Backman, J.T., & Niemi, M. (2018) Effects of Genetic Variants on Carboxylesterase 1 Gene Expression, and Clopidogrel Pharmacokinetics and Antiplatelet Effects. Basic & Clinical Pharmacology & Toxicology, 122(3), 341-345. DOI
  3. Imai, T., & Ohura, K. (2010) The Role of Intestinal Carboxylesterase in the Oral Absorption of Prodrugs. Current Drug Metabolism, 11(9), 793-805. DOI
  4. Jana, S., Mandlekar, S., & Marathe, P. (2010) Prodrug Design to Improve Pharmacokinetic and Drug Delivery Properties: Challenges to the Discovery Scientists. Current Medicinal Chemistry, 17(32), 3874-3908. DOI
  5. Geshi, E., Kimura, T., Yoshimura, M., Suzuki, H., Koba, S., Sakai, T., Saito, T., Koga, A., Muramatsu, M., & Katagiri, T. (2005) A single nucleotide polymorphism in the carboxylesterase gene is associated with the responsiveness to imidapril medication and the promoter activity. Hypertension Research, 28(9), 719-725. DOI
  6. Staudinger, J.L., Xu, C., Cui, Y.J., & Klaassen, C.D. (2010) Nuclear receptor-mediated regulation of carboxylesterase expression and activity. Expert Opinion on Drug Metabolism & Toxicology, 6(3), 261-271. DOI
  7. Imai, T. (2006) Human Carboxylesterase Isozymes: Catalytic Properties and Rational Drug Design. Drug Metabolism and Pharmacokinetics, 21(3), 173-185. DOI
  8. Sanghani, S.P., Sanghani, P.C., Schiel, M.A., & Bosron, W.F. (2009) Human Carboxylesterases: An Update on CES1, CES2 and CES3. Protein & Peptide Letters, 16(10), 1207-1214. DOI
  9. Taketani, M., Shii, M., Ohura, K., Ninomiya, S., & Imai, T. (2007) Carboxylesterase in the liver and small intestine of experimental animals and human. Life Sciences, 81(11), 924-932. DOI
  10. Satoh, T., & Hosokawa, M. (2006) Structure, function and regulation of carboxylesterases. Chemico-Biological Interactions, 162(3), 195-211. DOI
  11. Potter, P., & Wadkins, R. (2006) Carboxylesterases - Detoxifying Enzymes and Targets for Drug Therapy. Current Medicinal Chemistry, 13(9), 1045-1054. DOI
  12. Hatfield, M.J., & Potter, P.M. (2011) Carboxylesterase inhibitors. Expert Opinion on Therapeutic Patents, 21(8), 1159-1171. DOI
  13. Redinbo, M.R., & Potter, P.M. (2005) Keynote review: Mammalian carboxylesterases: From drug targets to protein therapeutics. Drug Discovery Today, 10(5), 313-325. DOI
  14. Rook, E., Huitema, A., Brink, W., Ree, J., & Beijnen, J. (2006) Pharmacokinetics and Pharmacokinetic Variability of Heroin and its Metabolites: Review of the Literature. Current Clinical Pharmacology, 1(1), 109-118. DOI
  15. Rudakova, E.V., Makhaeva, G.F., Galenko, T.G., Aksinenko, A.Y., Sokolov, V.B., & Martynov, I.V. (2013) A new selective inhibitor of mouse blood plasma carboxylesterase. Doklady Biochemistry and Biophysics, 449, 87-89. DOI
  16. Makhaeva, G.F., Rudakova, E.V., Aksinenko, A.Y., Sokolov, V.B., & Richardson, R.J. (2016) (O,O-Dibutyl)-O-1-trifluoromethyl-2,2,2-trifluoroethyl phosphate (BFP): A selective inhibitor of mouse plasma carboxylesterase. Phosphorus, Sulfur, and Silicon and the Related Elements, 191(11-12), 1589-1590. DOI
  17. Mukhamadieva, G.R., Boltneva, N.P., Galenko, T.G., Sokolov, V.B., Epishina, T.A., & Makhaeva, G.F. (2012) Synthesis and biological activity of O-carbamoylated 1,1,1,3,3,3-hexafluoroisopropanols as new specific inhibitors of carboxylesterase. Pharmaceutical Chemistry Journal, 46(8), 461-464. DOI
  18. Wadkins, R.M., Hyatt, J.L., Yoon, K.J., Morton, C.L., Lee, R.E., Damodaran, K., Beroza, P., Danks, M.K., & Potter, P.M. (2004) Discovery of novel selective inhibitors of human intestinal carboxylesterase for the amelioration of irinotecan-induced diarrhea: synthesis, quantitative structure-activity relationship analysis, and biological activity. Molecular Pharmacology, 65(6), 1336-1343. DOI
  19. Hicks, L.D., Hyatt, J.L., Stoddard, S., Tsurkan, L., Edwards, C.C., Wadkins, R.M., & Potter, P.M. (2009) Improved, selective, human intestinal carboxylesterase inhibitors designed to modulate 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (Irinotecan; CPT-11) toxicity. Journal of Medicinal Chemistry, 52(12), 3742-3752. DOI
  20. Wadkins, R.M., Hyatt, J.L., Wei, X., Yoon, K.J., Wierdl, M., Edwards, C.C., Morton, C.L., Obenauer, J.C., Damodaran, K., Beroza, P., Danks, M.K., & Potter, P.M. (2005) Identification and characterization of novel benzil (diphenylethane-1,2-dione) analogues as inhibitors of mammalian carboxylesterases. Journal of Medicinal Chemistry, 48(8), 2906-2915. DOI
  21. Hyatt, J.L., Moak, T., Hatfield, M.J., Tsurkan, L., Edwards, C.C., Wierdl, M., Danks, M.K., Wadkins, R.M., & Potter, P.M. (2007) Selective inhibition of carboxylesterases by isatins, indole-2,3-diones. Journal of Medicinal Chemistry, 50(8), 1876-1885. DOI
  22. Young, B.M., Hyatt, J.L., Bouck, D.C., Chen, T., Hanumesh, P., Price, J., Boyd, V.A., Potter, P.M., & Webb, T.R. (2010) Structure-activity relationships of substituted 1-pyridyl-2-phenyl-1,2-ethanediones: potent, selective carboxylesterase inhibitors. Journal of Medicinal Chemistry, 53(24), 8709-8715. DOI
  23. McKay, M.J., Carroll, A.R., Quinn, R.J., & Hooper, J.N.A. (2002) 1,2-Bis(1H-indol-3-yl)ethane-1,2-dione, an Indole Alkaloid from the Marine Sponge Smenospongia sp. Journal of Natural Products, 65(4), 595-597. DOI
  24. Hatfield, M.J., Tsurkan, L.G., Hyatt, J.L., Edwards, C.C., Lemoff, A., Jeffries, C., Yan, B., & Potter, P.M. (2013) Modulation of esterified drug metabolism by tanshinones from Salvia miltiorrhiza ("Danshen"). Journal of Natural Products, 76(1), 36-44. DOI
  25. Zou, L.W., Li, Y.G., Wang, P., Zhou, K., Hou, J., Jin, Q., Hao, D.C., Ge, G.B., & Yang, L. (2016) Design, synthesis, and structure-activity relationship study of glycyrrhetinic acid derivatives as potent and selective inhibitors against human carboxylesterase 2. European Journal of Medicinal Chemistry, 112, 280-288. DOI
  26. Binder, R.J., Hatfield, M.J., Chi, L., & Potter, P.M. (2018) Facile synthesis of 1,2-dione-containing abietane analogues for the generation of human carboxylesterase inhibitors. European Journal of Medicinal Chemistry, 149, 79-89. DOI
  27. Hatfield, M.J., Chen, J., Fratt, E.M., Chi, L., Bollinger, J.C., Binder, R.J., Bowling, J., Hyatt, J.L., Scarborough, J., Jeffries, C., & Potter, P.M. (2017) Selective Inhibitors of Human Liver Carboxylesterase Based on a beta-Lapachone Scaffold: Novel Reagents for Reaction Profiling. Journal of Medicinal Chemistry, 60(4), 1568-1579. DOI
  28. Wang, D.D., Zou, L. W., Jin, Q., Hou, J., Ge, G.B., & Yang, L. (2017) Recent progress in the discovery of natural inhibitors against human carboxylesterases. Fitoterapia, 117, 84-95. DOI
  29. Weng, Z.M., Ge, G.B., Dou, T.Y., Wang, P., Liu, P.K., Tian, X.H., Qiao, N., Yu, Y., Zou, L.W., Zhou, Q., Zhang, W.D., & Hou, J. (2018) Characterization and structure-activity relationship studies of flavonoids as inhibitors against human carboxylesterase 2. Bioorganic Chemistry, 77, 320-329. DOI
  30. Wheelock, C.E., Severson, T.F., & Hammock, B.D. (2001) Synthesis of new carboxylesterase inhibitors and evaluation of potency and water solubility. Chemical Research in Toxicology, 14(12), 1563-1572. DOI
  31. Boltneva, N.P., Makhaeva, G.F., Kovaleva, N.V., Lushchekina, S.V., Burgart, Y.V., Shchegol'kov, E.V., Saloutin, V.I., & Chupakhin, O.N. (2015) Alkyl 2-arylhydrazinylidene-3-oxo-3-polyfluoroalkylpropionates as new effective and selective inhibitors of carboxylesterase. Dokllady Biochemistry Biophysics, 465, 381-385. DOI
  32. Shchegolkov, E.V., Burgart, Y.V., Khudina, O.G., Saloutin, V.I., & Chupakhin, O.N. (2010) 2-(Het)arylhydrazono-1,3-dicarbonyl compounds in organic synthesis. Russian Chemical Reviews, 79(1), 31-61. DOI
  33. Khudina, O.G., Burgart, Y.V., Shchegol’kov, E.V., Saloutin, V.I., Kazheva, O.N., Chekhlov, A.N., & D’yachenko, O.A. (2009) Steric structure of alkyl 2-aryl(hetaryl)hydrazono-3-fluoroalkyl-3-oxopropionates. Russian Journal of Organic Chemistry, 45(6), 801-809. DOI
  34. Burgart, Y.V., Fokin, A.S., Kuzueva, O.G., Chupakhin, O.N., & Saloutin, V.I. (1998) Synthesis of fluorinated 2(3)-arylhydrazones of 1,2,3-tri(1,2,3,4-tetra)carbonyl compounds and their heterocyclization reactions. Journal of Fluorine Chemistry, 92(2), 101-108. DOI
  35. Shchegol'kov, E.V., Makhaeva, G.F., Boltneva, N.P., Lushchekina, S.V., Serebryakova, O.G., Rudakova, E.V., Kovaleva, N.V., Burgart, Y.V., Saloutin, V.I., Chupakhin, O.N., Bachurin, S.O., & Richardson, R.J. (2017) Synthesis, molecular docking, and biological activity of polyfluoroalkyl dihydroazolo[5,1-c][1,2,4]triazines as selective carboxylesterase inhibitors. Bioorganic & Medicinal Chemistry, 25(15), 3997-4007. DOI
  36. Shchegol’kov, E.V., Sadchikova, E.V., Burgart, Y.V., & Saloutin, V.I. (2009) Synthesis and structure of 4-hydroxy-4-fluoroalkyl-1,4-dihydroimidazo[5,1-c][1,2,4]triazines. Russian Journal of Organic Chemistry, 45(4), 572-580. DOI
  37. Shchegol’kov, E.V., Sadchikova, E.V., Burgart, Y.V., & Saloutin, V.I. (2008) Alkyl 3-fluoroalkyl-3-oxopropionates in reactions with azolyldiazonium salts. Russian Chemical Bulletin, 57(3), 612-616. DOI
  38. Shchegol'kov, E.V., Ivanova, A.E., Burgart, Y.V., & Saloutin, V.I. (2013) A Convenient Approach to 4,7-Dihydrotetrazolo[5,1-c][1,2,4]triazine Synthesis. Journal of Heterocyclic Chemistry, 50(S1), E80-E86. DOI
  39. Heymann, E., & Krisch, K. (1967) Phosphoric acid-bis-(p-nitro-phenylester), a new inhibitor of microsomal carboxylesterases. Hoppe-Seyler's Zeitschrift Fur Physiologische Chemie, 348(6), 609-619.
  40. Tsvetkov, E.N., Degtyarev, A.N., Bovin, A.N., Makhaeva, G.F., Yankovskaya, V.L. (1985) Russian Federation Patent No. 1187444.
  41. Makhaeva, G.F., Rudakova, E.V., Aksinenko, A.Yu., Sokolov, V.B., Serebryakova, O.G., Galenko, T.G. (2011) Russian Federation Patent No. 2423132.
  42. Makhaeva, G.F., Boltneva, N.P., Mukhamadieva, G.R., Sokolov, V.B., Galenko, T.G. (2012) Russian Federation Patent No. 2449988.
  43. Székács, A., Bordás, B., & Hammock, B.D. (1992) Transition state analog enzyme inhibitors: Structure-activity relationships of trifluoromethyl ketones. In: Rational Approaches to Structure, Activity, and Ecotoxicology of Agrochemicals (Draber W. & Fujita T., eds.), (pp. 219-249). Boca Raton, Florida: CRC Press.
  44. Burgart, Y.V., Makhaeva, G.F., Shchegol’kov, E.V., Khudina, O.G., Boltneva, N.P., Serebryakova, O.G., Lushchekina, S.V., Saloutin, V.I., Bachurin, S.O., Chupakhin, O.N. (2015) Russian Federation Patent No. 2574291.