Skip to main content
Log in

Clinical Pharmacokinetics of Pravastatin

Mechanisms of Pharmacokinetic Events

  • Review Articles
  • Drug Disposition
  • Published:
Clinical Pharmacokinetics Aims and scope Submit manuscript

Abstract

Pravastatin, one of the 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors (statins) widely used in the management of hypercholesterolaemia, has unique pharmacokinetic characteristics among the members of this class. Many In vivo and In vitro human and animal studies suggest that active transport mechanisms are involved in the pharmacokinetics of pravastatin.

The oral bioavailability of pravastatin is low because of incomplete absorption and a first-pass effect. The drug is rapidly absorbed from the upper part of the small intestine, probably via proton-coupled carrier-mediated transport, and then taken up by the liver by a sodium-independent bile acid transporter. About half of the pravastatin that reaches the liver via the portal vein is extracted by the liver, and this hepatic extraction is mainly attributed to biliary excretion which is performed by a primary active transport mechanism. The major metabolites are produced by chemical degradation in the stomach rather than by cytochrome P450-dependent metabolism in the liver. The intact drug and its metabolites are cleared through both hepatic and renal routes, and tubular secretion is a predominant mechanism in renal excretion.

The dual routes of pravastatin elimination reduce the need for dosage adjustment if the function of either the liver or kidney is impaired, and also reduce the possibility of drug interactions compared with other statins, which are largely eliminated by metabolism. The lower protein binding than other statins weakens the tendency for displacement of highly protein-bound drugs. Although all statins show a hepatoselective disposition, the mechanism for pravastatin is different from that of the others. There is high uptake of pravastatin by the liver via an active transport mechanism, but not by other tissues because of its hydrophilicity, whereas the disposition characteristics of other statins result from high hepatic extraction because of high lipophilicity.

These pharmacokinetic properties of pravastatin may be the result of the drug being given in the pharmacologically active open hydroxy acid form and the fact that its hydrophilicity is markedly higher than that of other statins. The nature of the pravastatin transporters, particularly in humans, remains unknown at present. Further mechanistic studies are required to establish the pharmacokinetic-pharmacodynamic relationships of pravastatin and to provide the optimal therapeutic efficacy for various types of patients with hypercholesterolaemia.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Table I
Table II
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Gagné C, Moorijani S, Brum D, et al. Heterozygous familial hypercholesterolemia and the relationship between plasma lipids, lipoproteinemias, clinical manifestations and ischemic heart disease in men and women. Atherosclerosis 1979; 34: 13–24.

    Article  PubMed  Google Scholar 

  2. Holme I. An analysis of randomized trials evaluating the effect of cholesterol reduction on total mortality and coronary heart disease incidence. Circulation 1990; 82: 1916–24.

    Article  PubMed  CAS  Google Scholar 

  3. Hoeg JM, Brewer Jr HB. 3-Hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors in the treatment of hypercholesterolemia. J Am Med Assoc 1987; 258: 3532–6.

    Article  CAS  Google Scholar 

  4. Grundy SM. HMG-CoA reductase inhibitors for treatment of hypercholesterolemia. N Engl J Med 1988; 319: 24–32.

    Article  PubMed  CAS  Google Scholar 

  5. Shepherd J, Cobbe SM, Ford I, et al. Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia. N Engl J Med 1995; 333: 1301–7.

    Article  PubMed  CAS  Google Scholar 

  6. Gotto AM. Lipid lowering, regression, and coronary events — a review of the interdisciplinary council on lipids and cardiovascular risk intervention, Seventh council meeting. Circulation 1993; 87: 1781–91.

    Article  Google Scholar 

  7. Dietschy JM, Wilson JD. Regulation of cholesterol metabolism 3. N Engl J Med 1970; 282: 1241–9.

    Article  PubMed  CAS  Google Scholar 

  8. Brown MS, Goldstein JL. A receptor-mediated pathway for cholesterol homeostasis. Science 1986; 232: 34–47.

    Article  PubMed  CAS  Google Scholar 

  9. Endo A. The discovery and development of HMG-Co A reductase inhibitors. J Lipid Res 1992; 33: 1569–82.

    PubMed  CAS  Google Scholar 

  10. Alberts AW. Discovery, biochemistry and biology of lovastatin. Am J Cardiol 1988; 62: 10J–5J.

    Article  PubMed  CAS  Google Scholar 

  11. Hoffman WF, Alberts AW, Anderson PS, et al. 3-Hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors 4: side chain ester derivatives of mevinolin. J Med Chem 1986; 29: 849–52.

    Article  PubMed  CAS  Google Scholar 

  12. Plosker GL, Wagstaff AJ. Fluvastatin: a review of its pharmacology and use in the management of hypercholesterolaemia. Drugs 1996; 51: 433–59.

    Article  PubMed  CAS  Google Scholar 

  13. Lea AP, McTavish D. Atorvastatin: a review of its pharmacology and therapeutic potential in the management of hyperlipidaemias. Drugs 1997; 53: 828–47.

    Article  PubMed  CAS  Google Scholar 

  14. Bischoff H, Angerbauer R, Bender J, et al. Cerivastatin: pharmacology of a novel synthetic and highly active HMG-CoA reductase inhibitor. Atherosclerosis 1997; 135: 119–30.

    Article  PubMed  CAS  Google Scholar 

  15. Komai T, Shigehara E, Tokui T, et al. Carrier-mediated uptake of pravastatin by rat hepatocytes in primary culture. Biochem Pharmacol 1992; 43: 667–70.

    Article  PubMed  CAS  Google Scholar 

  16. Ziegler K, Hummelsiep S. Hepatoselective carrier-mediated sodium-independent uptake of pravastatin and pravastatin-lactone. Biochim Biophys Acta 1993; 1153: 23–33.

    Article  PubMed  CAS  Google Scholar 

  17. Yamazaki M, Suzuki H, Hanano M, et al. Na+-independent multispecific anion transporter mediates active transport of pravastatin into rat liver. Am J Physiol 1993; 264: G36–44.

    PubMed  CAS  Google Scholar 

  18. Komai T, Kawai K, Tokui T, et al. Disposition and metabolism of pravastatin sodium in rats, dogs and monkeys. Eur J Drug Metab Pharmacokinet 1992; 17: 103–13.

    Article  PubMed  CAS  Google Scholar 

  19. Hatanaka T, Honda S, Sasaki S, et al. Pharmacokinetic and pharmacodynamic evaluation for tissue-selective inhibition of cholesterol synthesis by pravastatin. J Pharmacokinet Biopharm 1998; 26: 329–47.

    PubMed  CAS  Google Scholar 

  20. Tsujita Y, Kuroda M, Shimada K, et al. CS-514, a competitive inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase: tissue-selective inhibition of cholesterol synthesis and hypolipidemic effect on various animal species. Biochim Biophys Acta 1986; 877: 50–60.

    Article  PubMed  CAS  Google Scholar 

  21. Koga T, Shimada Y, Kuroda M, et al. Tissue-selective inhibition of cholesterol synthesis in vivo by pravastatin sodium, a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor. Biochim Biophys Acta 1990; 1045: 115–20.

    Article  PubMed  CAS  Google Scholar 

  22. McTavish D, Sorkin EM. Pravastatin: a review of its pharmacological properties and therapeutic potential in hypercholesterolaemia. Drugs 1991; 42: 65–89.

    Article  PubMed  CAS  Google Scholar 

  23. Quion JAV, Jones PH. Clinical pharmacokinetics of pravastatin. Clin Pharmacokinet 1994; 27: 94–104.

    Article  PubMed  CAS  Google Scholar 

  24. Desager JP, Horsmans Y. Clinical pharmacokinetics of 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors. Clin Pharmacokinet 1996; 31: 348–71.

    Article  PubMed  CAS  Google Scholar 

  25. Lennernäs H, Fager G. Pharmacodynamics and pharmacokinetics of the HMG-CoA reductase inhibitors: similarities and differences. Clin Pharmacokinet 1997; 32: 403–25.

    Article  PubMed  Google Scholar 

  26. Moghadasian MH. Clinical pharmacology of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors. Life Sci 1999; 65: 1329–37.

    Article  PubMed  CAS  Google Scholar 

  27. Haruyama H, Kuwano H, Kinoshita T, et al. Structure elucidation of the bioactive metabolites of ML-236B (mevastatin) isolated from dog urine. Chem Pharm Bull 1986; 34: 1459–67.

    Article  PubMed  CAS  Google Scholar 

  28. Matsuoka T, Miyakoshi S, Tanzawa K, et al. Purification and characterization of cytochrome P-450sca from Streptomyces carbophilus: ML-236B (compactin) induces a cytochrome P-450sca in Streptomyces carbophilus that hydroxylates ML-236B to pravastatin sodium (CS-514), a tissue-selective inhibitor of 3-hydroxy-3-methylglutaryl-coenzyme-A reductase. Eur J Biochem 1989; 184: 707–13.

    Article  PubMed  CAS  Google Scholar 

  29. Mevalotin — Interview Form. Tokyo: Sankyo, 1991.

  30. Serajuddin ATM, Ranadive SA, Mahoney EM. Relative lipophilicities, solubilities, and structure-pharmacological considerations of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors pravastatin, lovastatin, mevastatin, and simvastatin. J Pharm Sci 1991; 80: 830–4.

    Article  PubMed  CAS  Google Scholar 

  31. Alberts AW, Chen J, Kuron G, et al. Mevinolin: a highly potent competitive inhibitor of hydroxymethylglutaryl-coenzyme A reductase and a cholesterol-lowering agent. Proc Natl Acad Sci U S A 1980; 77: 3957–61.

    Article  PubMed  CAS  Google Scholar 

  32. Pan HY, DeVault AR, Wang-Iverson D, et al. Comparative pharmacokinetics and pharmacodynamics of pravastatin and lovastatin. J Clin Pharmacol 1990; 30: 1128–35.

    PubMed  CAS  Google Scholar 

  33. Pan HY, Triscari J, DeVault AR, et al. Pharmacokinetic interaction between propranolol and the HMG-CoAreductase inhibitors pravastatin and lovastatin. Br J Clin Pharmacol 1991; 31: 665–70.

    Article  PubMed  CAS  Google Scholar 

  34. Lilja JJ, Kivistö KT, Neuvonen PJ. Grapefruit juice increases serum concentrations of atorvastatin and has no effect on pravastatin. Clin Pharmacol Ther 1999; 66: 118–27.

    PubMed  CAS  Google Scholar 

  35. Funke PT, Ivashkiv E, Arnold ME, et al. Determination of pravastatin sodium and its major metabolite in serum/plasma by capillary gas chromatography/negative ion chemical ionization mass spectrometry. Biomed Environ Mass Spectr 1989; 18: 904–9.

    Article  CAS  Google Scholar 

  36. Singhvi SM, Pan HY, Morrison RA, et al. Disposition of pravastatin sodium, a tissue-selective HMG-CoA reductase inhibitor, in healthy subjects. Br J Clin Pharmacol 1990; 29: 239–43.

    Article  PubMed  CAS  Google Scholar 

  37. Muramatsu S, Takasaki W, Uchiyama M, et al. Enzyme-linked immunosorbent assay of pravastatin, a HMG-CoA reductase inhibitor, in human plasma. J Immunoassay 1996; 17: 13–27.

    Article  PubMed  CAS  Google Scholar 

  38. Dumousseaux C, Muramatsu S, Takasaki W, et al. Highly sensitive and specific determination of pravastatin sodium in plasma by high-performance liquid chromatography with laser-induced fluorescence determination after immobilized antibody extraction. J Pharm Sci 1994; 83: 1630–6.

    Article  PubMed  CAS  Google Scholar 

  39. Kawabata K, Matsushima N, Sasahara K, et al. An automated method for the simultaneous detection of pravastatin and its main metabolite in human plasma by high-performance liquid chromatography/atmospheric pressure chemical ionization mass spectrometry. Biomed Chromatogr 1998; 12: 271–5.

    Article  PubMed  CAS  Google Scholar 

  40. Botti RE, Triscari J, Pan HY, et al. Concentration of pravastatin and lovastatin in cerebrospinal fluid in healthy subjects. Clin Neuropharmacol 1991; 14: 256–61.

    Article  PubMed  CAS  Google Scholar 

  41. Pan HY, Waclawski AP, Funke PT, et al. Pharmacokinetics of pravastatin in elderly versus young men and women. Ann Pharmacother 1993; 27: 1029–33.

    PubMed  CAS  Google Scholar 

  42. Pan HY, DeVault AR, Brescia D, et al. Effect of food on pravastatin pharmacokinetics and pharmacodynamics. Int J Clin Pharmacol Ther Toxicol 1993; 31: 291–4.

    PubMed  CAS  Google Scholar 

  43. Triscari J, Swanson BN, Willard DA, et al. Steady state serum concentrations of pravastatin and digoxin when given in combination. Br J Clin Pharmacol 1993; 36: 263–5.

    Article  PubMed  CAS  Google Scholar 

  44. Everett DW, Chando TJ, Didonato GC, et al. Biotransformation of pravastatin sodium in human. Drug Metab Dispos 1991; 19: 740–8.

    PubMed  CAS  Google Scholar 

  45. Pan HY, DeVault AR, Swites BJ, et al. Pharmacokinetics and pharmacodynamics of pravastatin alone and with cholestyramine in hypercholesterolemia. Clin Pharmacol Ther 1990; 48: 201–7.

    Article  PubMed  CAS  Google Scholar 

  46. Sasahara K, Kawabata K, Nakaya N, et al. Phase I study of CS-514, an inhibitor of HMG-CoA reductase II: pharmacokinetics of CS-514 in healthy volunteers [in Japanese]. Rinsho Iyaku 1988; 4: 45–65.

    Google Scholar 

  47. Sigurbjörnsson S, Kjartansdòttir T, Jòhannsson M, et al. Apharmacokinetic evaluation of pravastatin in middle-aged and elderly volunteers. Eur J Drug Metab Pharmacokinet 1998; 23: 13–8.

    Article  PubMed  Google Scholar 

  48. Arai M, Serizawa N, Terahara A, et al. Pravastatin sodium (CS-514): a novel cholesterol-lowering agent which inhibits HMG-CoA reductase. Annu Rep Sankyo Res Lab 1988; 40: 1–38.

    CAS  Google Scholar 

  49. Triscari J, O’Donnell D, Zinny M, et al. Gastrointestinal absorption of pravastatin in healthy subjects. J Clin Pharmacol 1995; 35: 142–4.

    PubMed  CAS  Google Scholar 

  50. Duggan DE, Chen IW, Bayne WF, et al. The physiological disposition of lovastatin. Drug Metab Dispos 1989; 17: 166–73.

    PubMed  CAS  Google Scholar 

  51. Tamai I, Takanaga H, Maeda H, et al. Proton-cotransport of pravastatin across intestinal brush-border membrane. Pharm Res 1995; 12: 1727–32.

    Article  PubMed  CAS  Google Scholar 

  52. Tsuji A, Takanaga H, Tamai I, et al. Transcellular transport of benzoic acid across Caco-2 cells by a pH-dependent and carrier-mediated transport mechanism. Pharm Res 1994; 11: 30–7.

    Article  PubMed  CAS  Google Scholar 

  53. Van Vliet AK, Van Thiel GCF, Huisman RH, et al. Different effects of 3-hydroxy-3-methylglutaryl-coenzyme Areductase inhibitors on sterol synthesis in various human cell types. Biochim Biophys Acta 1995; 1254: 105–11.

    Article  PubMed  Google Scholar 

  54. Germershausen JI, Hunt VM, Bostedor RG, et al. Tissue selectivity of the cholesterol-lowering agents lovastatin, simvastatin and pravastatin in rats in vivo. Biochem Biophys Res Commun 1989; 158: 667–75.

    Article  PubMed  CAS  Google Scholar 

  55. Sliskovic DR, Roth BD, Bocan TMA. Tissue selectivity of HMG-CoA reductase inhibitors. Drug News Perspect 1992; 5: 517–33.

    Google Scholar 

  56. Yamazaki M, Tokui T, Ishigami M, et al. Tissue-selective uptake of pravastatin in rats: contribution of a specific carrier-mediated uptake system. Biopharm Drug Dispos 1996; 17: 775–89.

    Article  PubMed  CAS  Google Scholar 

  57. Ishigami M, Tokui T, Komai T, et al. Evaluation of the uptake of pravastatin by perfused rat liver and primary cultured rat hepatocytes. Pharm Res 1995; 12: 1741–5.

    Article  PubMed  CAS  Google Scholar 

  58. Tsuji A, Saheki A, Tamai I, et al. Transport mechanism of 3-hydroxy-3-methylglutaryl coenzyme Areductase inhibitors at the blood-brain barrier. J Pharm Exp Ther 1993; 267: 1085–90.

    CAS  Google Scholar 

  59. Koga T, Fukuda K, Shimada Y, et al. Tissue selectivity of pravastatin sodium, lovastatin and simvastatin: the relationship between inhibition of de novo synthesis and active drug concentrations in liver, spleen and testis in rat. Eur J Biochem 1992; 209: 315–9.

    Article  PubMed  CAS  Google Scholar 

  60. Yamazaki M, Akiyama S, Nishigaki R, et al. Uptake is the rate-limiting step in the overall hepatic elimination of pravastatin at steady-state in rats. Pharm Res 1996; 13: 1559–64.

    Article  PubMed  CAS  Google Scholar 

  61. Niinuma K, Kato Y, Suzuki H, et al. Primary active transport of organic anions on bile canalicular membrane in humans. Am J Physiol 1999; 276: G1153–64.

    PubMed  CAS  Google Scholar 

  62. Pan H, Fleiss P, Moore L, et al. Excretion of pravastatin, an HMG-CoA reductase inhibitor, in breast milk of lactating women [abstract]. J Clin Pharmacol 1988; 28: 942.

    Google Scholar 

  63. Jacobsen W, Kirchner G, Hallensleben K, et al. Small intestinal metabolism of the 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitor lovastatin and comparison with pravastatin. J Pharmacol Exp Ther 1999; 291: 131–9.

    PubMed  CAS  Google Scholar 

  64. Kitazawa E, Tamura N, Iwabuchi H, et al. Biotransformation of pravastatin sodium (I): mechanisms of enzymic transformation and epimerization of an allylic hydroxy group of pravastatin sodium. Biochem Biophys Res Commun 1993; 192: 592–602.

    Article  Google Scholar 

  65. Ito MK. Effects of extensive and poor gastrointestinal metabolism on the pharmacodynamics of pravastatin. J Clin Pharmacol 1998; 38: 331–6.

    PubMed  CAS  Google Scholar 

  66. Jacobsen W, Kirchner G, Hallensleben K, et al. Comparison of cytochrome P-450-dependent mechanism and drug interactions of the 3-hydroxy-3-methylglutaryl-Co A reductase inhibitors lovastatin and pravastatin in the liver. Drug Metab Dispos 1999; 27: 173–9.

    PubMed  CAS  Google Scholar 

  67. Transon C, Leemann T, Dayer P. In vitro comparative inhibition profiles of major human drug metabolising cytochrome P450 isozymes (CYP2C9, CYP2D6, and CYP3A4) by HMG-CoA reductase inhibitors. Eur J Clin Pharmacol 1996; 50: 209–15.

    Article  PubMed  CAS  Google Scholar 

  68. Halstenson CE, Triscari J, DeVault A, et al. Single-dose pharmacokinetics of pravastatin and metabolites in patients with renal impairment. J Clin Pharmacol 1992; 32: 124–32.

    PubMed  CAS  Google Scholar 

  69. Gehr TWB, Sica DA, Slugg PH, et al. The pharmacokinetics of pravastatin in patients on chronic hemodialysis. Eur J Clin Pharmacol 1997; 53: 117–21.

    Article  PubMed  CAS  Google Scholar 

  70. Garnett WR. Interactions with hydroxymethyl-glutaryl-coenzyme A reductase inhibitors. Am J Health Syst Pharm 1995; 52: 1639–45.

    PubMed  CAS  Google Scholar 

  71. Smith HT, Jokubaitis LA, Troendle AJ, et al. Pharmacokinetics of fluvastatin and specific drug interactions. Am J Hypertens 1993; 6: 17–26.

    Google Scholar 

  72. Radulovic LL, Cilla DD, Posvar EL, et al. Effect of food on the bioavailability of atorvastatin, an HMG-Co A reductase inhibitor. J Clin Pharmacol 1995; 35: 990–4.

    PubMed  CAS  Google Scholar 

  73. Levy G, Gibaldi M, Procknal JA. Effect of an anticholinergic agent on riboflavin absorption in man. J Pharm Sci 1972; 61: 798–9.

    Article  PubMed  CAS  Google Scholar 

  74. Kantola T, Kivistö KT, Neuvonen PJ. Grapefruit juice greatly increases serum concentrations of lovastatin and lovastatin acid. Clin Pharmacol Ther 1998; 63: 397–402.

    Article  PubMed  CAS  Google Scholar 

  75. Lilja JJ, Kivistö KT, Neuvonen PJ. Grapefruit juice-simvastatin interaction: effect on serum concentrations of simvastatin, simvastatin acid, and HMG-CoA reductase inhibitors. Clin Pharmacol Ther 1998; 64: 477–83.

    Article  PubMed  CAS  Google Scholar 

  76. Pan HY. Clinical pharmacology of pravastatin, a selective inhibitor of HMG-CoA reductase inhibitors. Clin Pharmacol Ther 1998; 64: 477–83.

    Article  Google Scholar 

  77. Tobert SA, Shear CL, Chremos AN, et al. Clinical experience with lovastatin. Am J Cardiol 1990; 65: 23F–26F.

    Article  PubMed  CAS  Google Scholar 

  78. Lettieri J, Krol G, Mazzu A, et al. Lack of pharmacokinetic interaction between cerivastatin, a new HMG-CoA reductase inhibitor, and digoxin [abstract 109]. Atherosclerosis 1997; 130: S29.

    Article  Google Scholar 

  79. Yang BB, Smithers JA, Siedlik PH, et al. Atorvastatin pharma cokinetic interactions with other CYP3A4 substrates: erythromycin and ethinyl estradiol [abstract]. Pharm Res 1996; 13: S437.

    Google Scholar 

  80. Mück W, Ritter W, Frey R, et al. Influence of cholestyramine on the pharmacokinetics of cerivastatin. Int J Clin Pharmacol Ther 1997; 35: 250–4.

    PubMed  Google Scholar 

  81. Lewis RV, McDevitt DG. Adverse reactions and interactions with ß-adrenoceptor blocking drugs. Med Toxicol 1986; 1: 343–61.

    PubMed  CAS  Google Scholar 

  82. Neuvonen PJ, Kantola T, Kivistö KT. Simvastatin but not pravastatin is very susceptible to interaction with the CYP3A4 inhibitor itraconazole. Clin Pharmacol Ther 1998; 63: 332–41.

    Article  PubMed  CAS  Google Scholar 

  83. Azie NE, Brater DC, Becker PA, et al. The interaction of diltiazem with lovastatin and pravastatin. Clin Pharmacol Ther 1998; 64: 369–77.

    Article  PubMed  CAS  Google Scholar 

  84. Pan HY, Swanson BN, DeVault AR, et al. Antipyrine elimination is not affected by chronic administration of pravastatin (SQ 31,000), a tissue-selective HMG-CoA reductase inhibitor [abstract]. Clin Res 1988; 36: 368A.

    Google Scholar 

  85. Mück W, Ochmann K, Unger S, et al. Influence of erythromycin pre- and co-treatment on single-dose pharmacokinetics of the HMG-CoA reductase inhibitor cerivastatin. Eur J Clin Pharmacol 1998; 53: 469–73.

    Article  PubMed  Google Scholar 

  86. Regazzi MB, Iacona I, Compana C, et al. Altered disposition of pravastatin following concomitant drug therapy with cyclosporin A in transplant recipients. Transplant Proc 1993; 25: 2732–4.

    PubMed  CAS  Google Scholar 

  87. Olbricht C, Wanner C, Eisenhauer T, et al. Accumulation of lovastatin, but not pravastatin, in the blood of cyclosporinetreated kidney graft patients after multiple doses. Clin Pharmacol Ther 1997; 62: 311–21.

    Article  PubMed  CAS  Google Scholar 

  88. Arnadottir M, Eriksson LO, Thysell H, et al. Plasma concentration profiles of simvastatin 3-hydroxy-3-methyl-glutaryl-co-enzyme A reductase inhibitor activity in kidney transplant recipients with and without ciclosporin. Nephron 1993; 65: 410–3.

    Article  PubMed  CAS  Google Scholar 

  89. Goldberg R, Roth D. Evaluation of fluvastatin in the treatment of hypercholesterolemia in renal transplant recipients taking cyclosporine. Transplant 1996; 62: 1559–64.

    Article  CAS  Google Scholar 

  90. Mück W, Mai I, Fritsche L, et al. Increase in cerivastatin systemic exposure after single and multiple dosing in cyclosporinetreated kidney transplant recipients. Clin Pharmacol Ther 1999; 65: 251–61.

    Article  PubMed  Google Scholar 

  91. Transon C, Leemann T, Vogt N, et al. In vivo inhibition profile of cytochrome P450TB (CYP2C9) by (+/−) fluvastatin. Clin Pharmacol Ther 1995; 58: 412–7.

    Article  PubMed  CAS  Google Scholar 

  92. Christians U, Jacobsen W, Floren LC. Metabolism and drug interactions of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors in transplant patients: are the statins mechanistically similar? Pharmacol Ther 1998; 80: 1–34.

    Article  PubMed  CAS  Google Scholar 

  93. Corsini A, Maggi FM, Catapano AL. Pharmacology of competitive inhibitors of HMG-CoAreductase. Pharmacol Res 1995; 31: 9–27.

    Article  PubMed  CAS  Google Scholar 

  94. McNamara PJ. Interspecies scaling in pharmacokinetics. Drug Pharm Sci 1991; 48: 267–300.

    CAS  Google Scholar 

  95. Sugiyama Y. Reconstruction of drug dispositions in vivo from in vitro studies based on physiological pharmacokinetic modeling: from low-molecular weight drugs to polypeptide compounds [in Japanese]. Yakugaku Zasshi 1989; 109: 199–231.

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Tomomi Hatanaka.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hatanaka, T. Clinical Pharmacokinetics of Pravastatin. Clin Pharmacokinet 39, 397–412 (2000). https://doi.org/10.2165/00003088-200039060-00002

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003088-200039060-00002

Keywords

Navigation