DOI QR코드

DOI QR Code

Dissolution and Duodenal Permeation Characteristics of Lovastatin from Bile Salt Solid Dispersions

담즙산염과의 고체분산체로부터 로바스타틴의 용출 및 십이지장 점막 투과 특성

  • Published : 2009.04.27

Abstract

Although lovastatin (LS) is widely used in the treatment of hypercholesterolemia, its bioavailability is known to be around 5%. This study was aimed to increase the solubility and dissolution-permeation rates of LS using solid dispersions (SDs) with bile salts. The solubilities of LS in water, aqueous bile salt solutions and non-aqueous vehicles were determined, and effects of bile salts on the cellulose or duodenal permeation of LS from SDs were evaluated using a horizontal permeation system. SDs were prepared at various ratios of LS to carriers, such as sodium deoxycholate (SDC), sodium glycocholate (SGC) and/or 2-hydroxypropyl-$\beta$-cyclodextrin (HPCD). The addition of bile salts (25 mM) in water increased markedly the solubility of LS by the micellar solubilization. Some non-aqueous vehicles were effective in solubilizing LS. From differential scanning calorimetric studies, it was found that the crystallinity of LS in SDs disappeared, indicating a formation of amorphous state. The SDs showed markedly enhanced dissolution compared with those of their physical mixtures (PMs) and drug alone. In the dissolution-permeation studies using a cellulose membrane, the donor and receptor solutions were maintained as a sink condition using pH 7.0 phosphate buffer containing 0.05% sodium lauryl sulfate (SLS). The flux of LS alone was nearly same as that of LS-SDC-HPCD (1:3:6) PM. However, the flux of LS-SDC-HPCD (1:3:6) SD slightly increased compared with drug alone and PM, suggesting that entrapment of LS in micelles does not significantly hinder the permeation across cellulose membrane. In the dissolution-duodenal permeation studies using a LS-HPCD-SDC (1:3:6) SD, the addition of various bile salts in donor solutions (25 mM) enhanced the permeation of LS markedly, and the fluxes were found to be $0.69{\pm}0.41$, $0.87{\pm}0.51$, $0.84{\pm}0.46$, $0.47{\pm}0.17$ and $0.68{\pm}0.32{\mu}g/cm^2/hr$ for sodium cholate (SC), SDC, SGC, sodium taurodeoxycholate (STDC) and sodium taurocholate (STC), respectively. The stepwise increase of donor SGC concentration increased the flux dose-dependently. From the relationship of donor SGC concentration and flux, the concentration of SGC initiating the permeation across the duodenal mucosa was calculated to be 11.1 mM, which is nearly same as the critical micelle concentration (CMC, 11.6 mM) of SGC. However, with no addition of bile salts and below CMC, the permeation was very limited and irratic, indicating that LS itself is very poor permeable. Higher protions of bile salt in SD such as LS-SDC or LS-SGC (1 : 49 and 1 : 69) showed highly promoted fluxes. In conclusion, SD systems with bile salts, which may form their micelles in intestinal fluids, might be a promising means for providing enhanced dissolution and intestinal permeation of practically insoluble and non-absorbable LS.

Keywords

References

  1. A.W. Alberts, Discovery biochemistry and biology of lovastatin, Am. J. Cardiol., 62, J10–J15 (1998). https://doi.org/10.1016/0002-9149(88)90002-1
  2. A. Corsini, S. Bellosta, R. Baetta, R. Fumagalli, R. Paoletti and F. Bernini, New insights into the pharmacodynamic and pharmacokinetic properties of statins, Pharmacol. Ther., 84, 413-428 (1999). https://doi.org/10.1016/S0163-7258(99)00045-5
  3. Y. Shitara and Y. Sugiyama, Pharmacokinetic and pharmacodynamic alterations of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors: drug-drug interactions and interindividual differences in transporter and metabolic enzyme functions, Pharmacol. Ther., 112, 71-105 (2006). https://doi.org/10.1016/j.pharmthera.2006.03.003
  4. R.H. Bradford, C.L. Shear, A.N. Chremos, C.A. Dujovne, F.A. Franklin, R.B. Grillo, J. Higgins, A. Langendörfer, D.T. Nash, J.L. Pool and H. Schnaper, Expanded clinical evaluation of lovastatin (EXCEL) study results: Two-year efficacy and safety follow-up, Am. J. Cardiol., 74, 667-673 (1994). https://doi.org/10.1016/0002-9149(94)90307-7
  5. J.R. Crouse III, P. Lukacsko and R. Niecestro, The Lovastatin Extended-Release Study Group, Dose response, safety and efficacy of an extended-release formulation of lovastatin in adults with hypercholesterolemia, Am. J. Cardiol., 89, 226-229 (2002). https://doi.org/10.1016/S0002-9149(01)02207-X
  6. M.H. Davidson, P. Lukacsko, J.X. Sun, G. Phillips, E. Walters, A. Sterman, R. Niecestro and L. Friedhoff, A multiple-dose pharmacodynamic, safety and pharmacokinetic comparison of extended- and immediate-release formulations of lovastatin, Clin. Ther., 24, 112-125 (2002). https://doi.org/10.1016/S0149-2918(02)85009-3
  7. R.W. Wang, P.H. Kari, A.Y. Lu, P.E. Thomas, F.P. Guengerich and K.P. Vyas, Biotranformation of lovastatin: IV. Identification of cytochrome P450 3A proteins as the major enzymes responsible for the oxidative metabolism of lovastatin in rat and human liver microsomes, Arch. Biochem. Biophys., 290, 355-361 (1991). https://doi.org/10.1016/0003-9861(91)90551-S
  8. T. Prueksartanont, R. Subramanian, X. Fang, B. Ma, Y. Qiu and J.H. Lin, Glucuronidation of statins in animals and humans: a novel mechanism of statin lactonization, Drug Metab. Dispos., 30, 505-512 (2002). https://doi.org/10.1124/dmd.30.5.505
  9. E. Wang, C.N. Casciano, R.P. Crement and W.W. Johnson, HMG-CoA reductase inhibitors (statins) characterized as direct inhibitors of P-glycoprotein, Pharm. Res., 18, 800-806 (2001). https://doi.org/10.1023/A:1011036428972
  10. G.A. McClelland, R.J. Stubbs, J.A. Fix, S.A. Pogany and G.M., Zentner, Enhancement of 3-hydroxy-3-methylglutarylcoenzyme A (HMG-CoA) reductase inhibitor efficacy through administration of a controlled-porosity osmotic pump dosage form. Pharm. Res., 8, 873-876 (1991). https://doi.org/10.1023/A:1015899328105
  11. B.K. Kang, B.Y. Yoon, K.S. Seo, S.Y. Jeung, J.H. Kil, G. Khang, H.B. Lee and S.H. Cho, Preparation of solid dosage form containing SMEDDS of simvastatin by microencapsulation. J. Kor. Pharm. Sci., 33, 121-127 (2003).
  12. G.L. Amidon, H. Lennernas, V.P. Shah and J.R. Crison, A theoretical basis for biopharmaceutical drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability, Pharm. Res., 12, 413-420 (1995). https://doi.org/10.1023/A:1016212804288
  13. M.V.S. Varma, S. Khandavilli, Y. Ashokraj, A. Jain, A. Dhanikula, A. Sood, N.S. Thomas, O.P. Pillai, P. Sharma, R. Gandhi, S. Agrawal, V. Nair and R. Panchagnula, Biopharmaceutic classification system: a scientific framework for pharmacokinetic optimization in drug research, Curr. Drug Metab., 5, 375-388 (2004). https://doi.org/10.2174/1389200043335423
  14. P. Sharma, M.V.S. Varma, H.P.S. Chawla and R. Panchagnula, Absorption enhancement, mechanistic and toxicity studies of medium chain fatty acids, cyclodextrins and bile salts as peroral absorption enhancers, Il Pharmaco, 60, 884-893 (2005).
  15. H. Sugioka and Y. Moroi, Micelle formation of sodium cholate and solubilization into the micelle, Biochim. Biophys. Acta, 1394, 99-110 (1998). https://doi.org/10.1016/S0005-2760(98)00090-3
  16. R. Ninomiya, K. Matsuoka and Y. Moroi, Micelle formation of sodium deoxycholate and solubilization into the micelles: comparison with other unconjugated bile salts, Biochem. Biophys. Acta, 1634, (2003). https://doi.org/10.1016/j.bbalip.2003.09.003
  17. S. Reis, C.G. Moutinho, C. Matos, B. de Castro, P. Gameiro and J.J.F.C. Lima, Noninvasive methods to determine the critical micelle concentration of some bile acid salts, Anal. Biochem., 334, 117-126 (2004). https://doi.org/10.1016/j.ab.2004.07.017
  18. U. Subuddhi and A.K. Mishra, Micellization of bile salts in aqueous medium: a fluorescence study, Colloids Surf. B: Biointerfaces, 57, 102-107 (2007). https://doi.org/10.1016/j.colsurfb.2007.01.009
  19. A. Coello, F. Meijide, E.R. Ninez and J.V. Tato, Aggregation behavior of bile salts in aqueous solutions, J. Pharm. Sci., 85, 9-15 (1996). https://doi.org/10.1021/js950326j
  20. K. Matsuoka, M. Maeda and Y. Moroi, Micelle formation of sodium glyco- and taurocholates and sodium glyco- and taurodeoxycholates and solubilization of cholesterol into their micelles, Colloids Surf. B: Biointerfaces, 32, 87-95 (2003). https://doi.org/10.1016/S0927-7765(03)00148-6
  21. K. Matsuoka, M. Suzuki, C. Honda, K. Endo and Y. Moroi, Micellization of conjugated chenodeoxy- and ursodeoxycholates and solubilization of cholesterol into their micelles: comparison with other four conjugated bile salts species, Chem. Phys. Lipids, 139, 1-10 (2006). https://doi.org/10.1016/j.chemphyslip.2005.08.006
  22. N. Yoshida, Y. Moroi, R. Humphry-Baker and M. Graetzel, Dynamics for solubilization of naphthalene and pyrene into n-decyltrimethylammonium perfluorocarboxylate micelles, J. Phys. Chem. A, 106, 3991-3997 (2002). https://doi.org/10.1021/jp0138072
  23. N.Y. Lee and I.K. Chun, Enhanced dissolution of simvastatin by solid dispersion with water-soluble carriers, Dongduk Pharm. Res., 12, 1-12 (2008).
  24. S. Gould and R.C. Scott, 2-Hydroxypropyl-ß-cyclodextrin (HP-ß-CD): a toxicological review, Food Chem. Toxicol., 43, 1451-1459 (2005). https://doi.org/10.1016/j.fct.2005.03.007
  25. X. Tan and S. Lindenbaum, Studies on complexation between ß-cyclodextrin and bile salts, Int. J. Pharm., 74, 127-1353 (1991). https://doi.org/10.1016/0378-5173(91)90229-H
  26. S. Comini, P. Olivier, M. Riottot and D. Duhamel, Interaction of ß-cyclodextrin with bile acids and their competition with vitamins A and D3 as determined by 1HNMR spectrometry, Clin. Chem. Acta, 228, 181-194 (1994). https://doi.org/10.1016/0009-8981(94)90288-7
  27. M.A. Vandelli, G. Salvioli, A. Mocci, R. Panini, L. Malmusi and F. Forni, 2-Hydroxypropyl-ß-cyclodextrin complexation with ursodeoxycholic acid, Int. J. Pharm., 118, 77-83 (1995). https://doi.org/10.1016/0378-5173(94)00342-3
  28. F.G.J. Poelma, R. Breas and J.J. Tukker, Intestinal absorption of drugs. IV. the influence of taurocholate and L-cysteine on the barrier function of mucus, Int. J. Pharm., 64, 161-169 (1990). https://doi.org/10.1016/0378-5173(90)90265-6
  29. P.M. van Hasselt, G.E.P.J. Janssens, T.K. Slot, M. Van der Ham, T.C. Minderhoud, M. Talelli, L.M. Akkermans, C.J.F. Rijcken and C.F. van Nostrum, The influence of bile salts on the oral bioavailability of vitamin K encapsulated in polymeric micelles, J. Contr. Rel., 133, 161-168 (2009). https://doi.org/10.1016/j.jconrel.2008.09.089
  30. I.K. Chun, D.D. Kim, Y.J. Wang and Y.W. Chien, Permeation enhancement of methionine enkephalin by combinations of enzyme inhibitors and enhancers, S.T.P. Pharma Sci., 6, 148-156 (1996).
  31. M.A. Hammad and B.W. Müller, Increasing drug solubility by means of bile salt-phosphatidylcholine-based mixed micelles, Eur. J. Pharm. Biopharm., 46, 361-367 (1998). https://doi.org/10.1016/S0939-6411(98)00037-X
  32. L. Forsgren, Studies on the intestinal absorption of labelled fat-soluble vitamins (A, D, E, and K) via the thoracic-duct lymph in the absence of bile in man, Acta Chir. Scand. Suppl., 399, 1-29 (1969).
  33. L.A. Woollett, Y. Wang, D.D. Buckley, L. Yao, S. Chin and N. Granholm, Micellar solubilisation of cholesterol is essential for absorption in humans, Gut, 55, 197-204 (2006). https://doi.org/10.1136/gut.2005.069906
  34. A.F. Hofmann, Intestinal absorption of bile acids and biliary constituents: the intestinal component of the enterohepatic circulation and the integrated system. In: Physiology of the Gastrointestinal Tract (3rd ed., vol. 2), edited by Johnson L.R., Alpers, D.H., Jacobson, E.D and Walsh, J.H., New York, Raven, 1994, p. 1845-1865
  35. A.F. Hofmann and K.J. Mysels, Bile acid solubility and precipitation in vitro and in vivo: the role of conjugation, pH and Ca2+ ions, J. Lipid Res., 33 617-626 (1992).