DOI QR코드

DOI QR Code

In vitro and in vivo pharmacokinetic characterization of LMT-28 as a novel small molecular interleukin-6 inhibitor

  • Ahn, Sung-Hoon (College of Pharmacy, Kangwon National University) ;
  • Heo, Tae-Hwe (Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea) ;
  • Jun, Hyun-Sik (Department of Biotechnology and Bioinformatics, College of Science and Technology, Korea University) ;
  • Choi, Yongseok (School of Life Sciences and Biotechnology, Korea University)
  • Received : 2019.06.04
  • Accepted : 2019.07.31
  • Published : 2020.04.01

Abstract

Objective: Interleukin-6 (IL-6) is a T cell-derived B cell stimulating factor which plays an important role in inflammatory diseases. In this study, the pharmacokinetic properties of LMT-28 including physicochemical property, in vitro liver microsomal stability and an in vivo pharmacokinetic study using BALB/c mice were characterized. Methods: LMT-28 has been synthesized and is being developed as a novel therapeutic IL-6 inhibitor. The physicochemical properties and in vitro pharmacokinetic profiles such as liver microsomal stability and Madin-Darby canine kidney (MDCK) cell permeability assay were examined. For in vivo pharmacokinetic studies, pharmacokinetic parameters using BALB/c mice were calculated. Results: The logarithm of the partition coefficient value (LogP; 3.65) and the apparent permeability coefficient values (Papp; 9.7×10-6 cm/s) showed that LMT-28 possesses a moderate-high cell permeability property across MDCK cell monolayers. The plasma protein binding rate of LMT-28 was 92.4% and mostly bound to serum albumin. The metabolic half-life (t1/2) values of LMT-28 were 15.3 min for rat and 21.9 min for human at the concentration 1 μM. The area under the plasma drug concentration-time curve and Cmax after oral administration (5 mg/kg) of LMT-28 were 302±209 h·ng/mL and 137±100 ng/mL, respectively. Conclusion: These data suggest that LMT-28 may have good physicochemical and pharmacokinetic properties and may be a novel oral drug candidate as the first synthetic IL-6 inhibitor to ameliorate mammalian inflammation.

Keywords

References

  1. Venkatesh S, Lipper RA. Role of the development scientist in compound lead selection and optimization. J Pharm Sci 2000;89:145-54. https://doi.org/10.1002/(SICI)1520-6017(200002)89:2<145::AID-JPS2>3.0.CO;2-6
  2. Kerns EH, Di L. Pharmaceutical profiling in drug discovery. Drug Discov Today 2003;8:316-23. https://doi.org/10.1016/S1359-6446(03)02649-7
  3. Han SY, Lee CO, Ahn SH, et al. Evaluation of a multi-kinase inhibitor KRC-108 as an anti-tumor agent in vitro and in vivo. Invest New Drugs 2012;30:518-23. https://doi.org/10.1007/s10637-010-9584-2
  4. Kerns EH, Di L. Physicochemical profiling: overview of the screens. Drug Discov Today Technol 2004;1:343-8. https://doi.org/10.1016/j.ddtec.2004.08.011
  5. Li AP. Screening for human ADME/Tox drug properties in drug discovery. Drug Discov Today 2001;6:357-66. https://doi.org/10.1016/S1359-6446(01)01712-3
  6. Scheller J, Garbers C, Rose-John S. Interleukin-6: from basic biology to selective blockade of pro-inflammatory activities. Semin Immunol 2014;26:2-12. https://doi.org/10.1016/j.smim.2013.11.002
  7. Hunter CA, Jones SA. IL-6 as a keystone cytokine in health and disease. Nat Immunol 2015;16:448-57. https://doi.org/10.1038/ni.3153
  8. Huang CM, Lee TT. Immunomodulatory effects of phytogenics in chickens and pigs - A review. Asian-Australas J Anim Sci 2018;31:617-27. https://doi.org/10.5713/ajas.17.0657
  9. Zhao S, Pang Y, Zhao X, Du W, Hao H, Zhu H. Detrimental effects of lipopolysaccharides on maturation of bovine oocytes. Asian-Australas J Anim Sci 2019;32:1112-21. https://doi.org/10.5713/ajas.18.0540
  10. Kumari N, Dwarakanath BS, Das A, Bhatt AN. Role of interleukin-6 in cancer progression and therapeutic resistance. Tumour Biol 2016;37:11553-72. https://doi.org/10.1007/s13277-016-5098-7
  11. Yao X, Huang J, Zhong H, et al. Targeting interleukin-6 in inflammatory autoimmune diseases and cancers. Pharmacol Ther 2014;141:125-39. https://doi.org/10.1016/j.pharmthera.2013.09.004
  12. Wolf J, Rose-John S, Garbers C. Interleukin-6 and its receptors: a highly regulated and dynamic system. Cytokine 2014;70: 11-20. https://doi.org/10.1016/j.cyto.2014.05.024
  13. Leu CM, Wong FH, Chang C, Huang SF, Hu CP. Interleukin-6 acts as an antiapoptotic factor in human esophageal carcinoma cells through the activation of both STAT3 and mitogen-activated protein kinase pathways. Oncogene 2003;22:7809-18. https://doi.org/10.1038/sj.onc.1207084
  14. Jones SA, Scheller J, Rose-John S. Therapeutic strategies for the clinical blockade of IL-6/gp130 signaling. J Clin Invest 2011;121:3375-83. https://doi.org/10.1172/JCI57158
  15. Hong SS, Choi JH, Lee SY, et al. A novel small-molecule inhibitor targeting the IL-6 receptor beta subunit, glycoprotein 130. J Immunol 2015;195:237-45. https://doi.org/10.4049/jimmunol.1402908
  16. Baka E, Comer JEA, Takacs-Novak K. Study of equilibrium solubility measurement by saturation shake-flask method using hydrochlorothiazide as model compound. J Pharm Biomed Anal 2008;46:335-41. https://doi.org/10.1016/j.jpba.2007.10.030
  17. Zhou L, Yang L, Tilton S, Wang J. Development of a high throughput equilibrium solubility assay using miniaturized shake-flask method in early drug discovery. J Pharm Sci 2007;96:3052-71. https://doi.org/10.1002/jps.20913
  18. Park JS, Kim MS, Song JS, et al. Dose-independent pharmacokinetics of a new peroxisome proliferator-activated receptor-gamma agonist, KR-62980, in Sprague-Dawley rats and ICR mice. Arch Pharm Res 2011;34:2051-8. https://doi.org/10.1007/s12272-011-1207-8
  19. Obach RS. Prediction of human clearance of twenty-nine drugs from hepatic microsomal intrinsic clearance data: An examination of in vitro half-life approach and nonspecific binding to microsomes. Drug Metab Dispos 1999;27:1350-9.
  20. Davies B, Morris T. Physiological parameters in laboratory animals and humans. Pharm Res 1993;10:1093-5. https://doi.org/10.1023/A:1018943613122
  21. van de Waterbeemd H, Testa B. Drug bioavailability: estimation of solubility, permeability, absorption and bioavailability. Weinheim, Germany: Wiley-VCH; 2009.
  22. Lipinski CA. Drug-like properties and the causes of poor solubility and poor permeability. J Pharmacol Toxicol Methods 2000;44:235-49. https://doi.org/10.1016/S1056-8719(00)00107-6
  23. Lipinski CA. Lead- and drug-like compounds: the rule-of-five revolution. Drug Discov Today Technol 2004;1:337-41. https://doi.org/10.1016/j.ddtec.2004.11.007
  24. Thapa RK, Choi HG, Kim JO, Yong CS. Analysis and optimization of drug solubility to improve pharmacokinetics. J Pharm Invest 2017;47:95-110. https://doi.org/10.1007/s40005-016-0299-z
  25. Nelson DR, Zeldin DC, Hoffman SM, Maltais LJ, Wain HM, Nebert DW. Comparison of cytochrome P450 (CYP) genes from the mouse and human genomes, including nomenclature recommendations for genes, pseudogenes and alternative-splice variants. Pharmacogenetics 2004;14:1-18. https://doi.org/10.1097/00008571-200401000-00001
  26. Azarara M, Afrasibirad A, Farzamikia N, Alijani A, Sakhinia E. The effect of GGCX and CYP4F2 gene polymorphisms in genotype-guided dosing of warfarin in patients with a history of cardiac surgery. J Pharm Invest 2017;47:349-355. https://doi.org/10.1007/s40005-016-0295-3
  27. Chen LL, Yao J, Yang JB, Yang J. Predicting MDCK cell permeation coefficients of organic molecules using membrane-interaction QSAR analysis. Acta Pharmacol Sin 2005;26:1322-33. https://doi.org/10.1111/j.1745-7254.2005.00166.x
  28. Zahner D, Alber J, Petzinger E. Cloning and heterologous expression of the ovine (Ovis aries) P-glycoprotein (Mdr1) in Madin-Darby canine kidney (MDCK) cells. J Vet Pharmacol Ther 2010;33:304-11. https://doi.org/10.1111/j.1365-2885. 2009.01141.x