Skip to main content
Log in

Pharmacokinetics of Melagatran and the Effect on Ex Vivo Coagulation Time in Orthopaedic Surgery Patients Receiving Subcutaneous Melagatran and Oral Ximelagatran

A Population Model Analysis

  • Original Research Article
  • Published:
Clinical Pharmacokinetics Aims and scope Submit manuscript

Abstract

Objective: Ximelagatran, an oral direct thrombin inhibitor, is rapidly bioconverted to melagatran, its active form. The objective of this population analysis was to characterise the pharmacokinetics of melagatran and its effect on activated partial thromboplastin time (APTT), an ex vivo measure of coagulation time, in orthopaedic surgery patients sequentially receiving subcutaneous melagatran and oral ximelagatran as prophylaxis for venous thromboembolism. To support the design of a pivotal dose-finding study, the impact of individualised dosage based on bodyweight and calculated Creatinine clearance was examined.

Design and methods: Pooled data obtained in three small dose-guiding studies were analysed. The patients received twice-daily administration, with either subcutaneous melagatran alone or a sequential regimen of subcutaneous melagatran followed by oral ximelagatran, for 8–11 days starting just before initiation of surgery. Nonlinear mixed-effects modelling was used to evaluate rich data of melagatran pharmacokinetics (3326 observations) and the pharmacodynamic effect on APTT (2319 observations) in samples from 216 patients collected in the three dose-guiding trials. The pharmacokinetic and pharmacodynamic models were validated using sparse data collected in a subgroup of 319 patients enrolled in the pivotal dose-finding trial. The impact of individualised dosage on pharmacokinetic and pharmacodynamic variability was evaluated by simulations of the pharmacokinetic-pharmacodynamic model.

Results: The pharmacokinetics of melagatran were well described by a one-compartment model with first-order absorption after both subcutaneous melagatran and oral ximelagatran. Melagatran clearance was correlated with renal function, assessed as calculated Creatinine clearance. The median population clearance (creatinine clearance 70 mL/min) was 5.3 and 22.9 L/h for the subcutaneous and oral formulations, respectively. The bioavailability of melagatran after oral ximelagatran relative to subcutaneous melagatran was 23%. The volume of distribution was influenced by bodyweight. For a patient with a bodyweight of 75kg, the median population estimates were 15.5 and 159L for the subcutaneous and oral formulations, respectively. The relationship between APTT and melagatran plasma concentration was well described by a power function, with a steeper slope during and early after surgery but no influence by any covariates. Simulations demonstrated that individualised dosage based on Creatinine clearance or bodyweight had no clinically relevant impact on the variability in melagatran pharmacokinetics or on the effect on APTT.

Conclusions: The relatively low impact of individualised dosage on the pharmacokinetic and pharmacodynamic variability of melagatran supported the use of a fixed-dose regimen in the studied population of orthopaedic surgery patients, including those with mild to moderate renal impairment.

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.

Table I
Fig. 1
Table II
Table III
Fig. 2
Fig. 3
Fig. 4
Table IV
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Weite JI, Hirsh J. New anticoagulant drags. Chest 2001; 119 (1 Suppl.): S95–107

    Article  Google Scholar 

  2. Elg M, Gustafsson D, Deinum J. The importance of enzyme kinetics for the effect of thrombin inhibitors in a rat model of arterial thrombosis. Thromb Haemost 1997; 78: 1286–92

    PubMed  CAS  Google Scholar 

  3. Elg M, Gustafsson D, Carlsson S. Antithrombotic effects and bleeding time of thrombin inhibitors and warfarin in the rat. Thromb Res 1999; 94: 187–97

    Article  PubMed  CAS  Google Scholar 

  4. Gustafsson D, Antonsson T, Bylund R, et al. Effects of melagatran, a new low-molecular-weight thrombin inhibitor, on thrombin and fibrinolytic enzymes. Thromb Haemost 1998; 79: 110–8

    PubMed  CAS  Google Scholar 

  5. Mehta JL, Chen L, Nichols WW, et al. Melagatran, an oral active-site inhibitor of thrombin, prevents or delays formation of electrically induced occlusive thrombus in the canine coronary artery. J Cardiovasc Pharmacol 1998; 31: 345–51

    Article  PubMed  CAS  Google Scholar 

  6. Eriksson BI, Carlsson S, Halvarsson M, et al. Antithrombotic effect of two low molecular weight thrombin inhibitors and a low molecular weight heparin in a caval vein thrombosis model in the rat. Thromb Haemost 1997; 78: 1404–7

    PubMed  CAS  Google Scholar 

  7. Mattsson C, Björkman J-A, Ulvinge J-C. Melagatran, hiradin and heparin as adjuncts to tissue-type Plasminogen activator in a canine model of coronary artery thrombolysis. Fibrinolysis Proteolysis 1997; 11: 121–8

    Article  CAS  Google Scholar 

  8. Sarich TC, Eriksson UG, Mattsson C, et al. Inhibition of thrombin generation by the oral direct thrombin inhibitor ximelagatran in shed blood from healthy male subjects. Thromb Haemost 2002; 87: 300–5

    PubMed  CAS  Google Scholar 

  9. Eriksson BI, Ögren M, Eriksson UG, et al. Prophylaxis of venous thromboembolism with subcutaneous melagatran in total hip or total knee replacement: results from Phase II studies. Thromb Res 2002; 105: 371–8

    Article  PubMed  CAS  Google Scholar 

  10. Eriksson BI, Arfwidsson A-C, Frison L, et al. A dose-ranging study of the oral direct thrombin inhibitor, ximelagatran, and its subcutaneous form, melagatran, compared with dalteparin in the prophylaxis of thromboembolism after hip or knee replacement: METHRO I. MElagatran for THRombin inhibition in Orthopaedic surgery. Thromb Haemost 2002; 87: 231–7

    PubMed  CAS  Google Scholar 

  11. Eriksson BI, Bergqvist D, Kälebo P, et al. The oral direct thrombin inhibitor, ximelagatran, and its active form, melagatran, in ascending doses compared with dalteparin for prevention of venous thromboembolism after total hip or total knee replacement: The METHRO II Study. Lancet 2002; 360: 1441–7

    Article  PubMed  CAS  Google Scholar 

  12. Eriksson H, Eriksson UG, Frison L, et al. Pharmacokinetics and pharmacodynamics of melagatran, a novel synthetic LMW thrombin inhibitor, in patients with acute DVT. Thromb Haemost 1999; 81: 358–63

    PubMed  CAS  Google Scholar 

  13. Petersen P, SPORTIF II and IV Investigators. A long-term follow-up of ximelagatran as an oral anticoagulant for the prevention of stroke and systemic embolism in patients with atrial fibrillation [abstract]. Blood 2001; 98 Suppl. 1: 706a

    Google Scholar 

  14. Bredberg U, Eriksson UG, Taure K, et al. Effect of melagatran, a novel direct thrombin inhibitor, in healthy volunteers following intravenous, subcutaneous and oral administration [abstract]. Blood 1999; 94 Suppl. 1: 28a

    Google Scholar 

  15. Gustafssson D, Nystrom J, Carlsson S, et al. The direct thrombin inhibitor melagatran and its oral prodrug H 376/95: intestinal absorption properties, biochemical and pharmacodynamic effects. Thromb Res 2001; 101: 171–81

    Article  Google Scholar 

  16. Eriksson UG, Bredberg U, Gislén K, et al. Pharmacokinetics and pharmacodynamics of ximelagatran, a novel oral direct thrombin inhibitor, in young healthy male subjects. Eur J Clin Pharmacol 2003; 59: 35–43

    PubMed  CAS  Google Scholar 

  17. Johansson LC, Frison L, Logren U, et al. The influence of age on the pharmacokinetics and pharmacodynamics of ximelagatran, an oral direct thrombin inhibitor. Clin Pharmacokinet 2003; 42: 381–92

    Article  PubMed  CAS  Google Scholar 

  18. Larsson M, Logren U, Ahnoff M, et al. Determination of melagatran, a novel direct thrombin inhibitor, in human plasma and urine by liquid chromatography: mass spectrometry. J Chromatogr B 2002; 766: 47–55

    Article  CAS  Google Scholar 

  19. Beal SL, Sheiner LB. NONMEM users guides. NONMEM Project Group. San Francisco: University of California, 1998

    Google Scholar 

  20. Cockcroft DW, Gault MH. Prediction of Creatinine clearance from serum creatinine. Nephron 1976; 16: 31–41

    Article  PubMed  CAS  Google Scholar 

  21. Cullberg M, Eriksson UG, Larsson M, et al. Population modelling of the effect of inogatran, a thrombin inhibitor, on ex vivo coagulation time (APTT) in healthy subjects and patients with coronary artery disease. Br J Clin Pharmacol 2001; 51: 71–9

    Article  PubMed  CAS  Google Scholar 

  22. Robson R, White H, Aylward P, et al. Bivalirudin pharmacokinetics and pharmacodynamics: effect of renal function, dose and gender. Clin Pharmacol Ther 2002, 9

Download references

Acknowledgements

Ulf G. Eriksson, Lars Frisson, Bengt Hamrén and David Gustafsson are employees of AstraZeneca. Jaap W. Mandema and Per Olsson Gisleskog were consultants for AstraZeneca. Mats O. Karlsson received research funding from AstraZeneca. Ulrika Wählby received research funding from and is presently an employee of AstraZeneca. Bengt I. Eriksson was principle investiagtor for the clinical studies and has declared no conflict of interest in connection with this paper.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ulf G. Eriksson.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Eriksson, U.G., Mandema, J.W., Karlsson, M.O. et al. Pharmacokinetics of Melagatran and the Effect on Ex Vivo Coagulation Time in Orthopaedic Surgery Patients Receiving Subcutaneous Melagatran and Oral Ximelagatran. Clin Pharmacokinet 42, 687–701 (2003). https://doi.org/10.2165/00003088-200342070-00006

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003088-200342070-00006

Keywords

Navigation