Pharmacokinetics of recombinant activated factor VII in trauma patients with severe bleeding

Thomas Klitgaard, Rene Tabanera y Palacios, Kenneth D Boffard, Philip T C Iau, Brian Warren, Sandro Rizoli, Rolf Rossaint, Yoram Kluger, Bruno Riou, NovoSeven Trauma Study Group, Thomas Klitgaard, Rene Tabanera y Palacios, Kenneth D Boffard, Philip T C Iau, Brian Warren, Sandro Rizoli, Rolf Rossaint, Yoram Kluger, Bruno Riou, NovoSeven Trauma Study Group

Abstract

Introduction: Recombinant activated factor VII (rFVIIa) has been used as adjunctive therapy in trauma patients with severe bleeding. However, its pharmacokinetics profile remains unknown.

Methods: In two placebo-controlled studies in patients with blunt and penetrating trauma, the pharmacokinetics of rFVIIa given at an initial dose of 200 microg x kg-1 after transfusion of eight red blood cell units, followed by additional doses of 100 microg x kg-1, one and three hours later, have been studied, based on the FVII coagulant activity assay. Both non-compartment and population pharmacokinetic analyses were performed. A two-compartment, population pharmacokinetic model was used to estimate a population profile for the rFVIIa dosing regimen. Data are population means (percent coefficient of variation (CV)).

Results: Based on the two-compartment population model, the estimated pharmacokinetic parameters were: clearance 40 (30% CV) ml x kg-1 x h-1; central volume of distribution 89 (32% CV) ml x kg-1; inter-compartmental clearance 24 ml x kg-1 x h-1; and peripheral compartment volume 31 ml x kg-1. Baseline FVII coagulant activity was estimated at 0.29 (39% CV) U x ml-1, initial half-life was 0.6 (34% CV) hours, and terminal half-life 2.4 (50% CV) hours. High intra- and inter-patient variability was noted in volume of distribution and clearance, which was in part correlated with the transfusion requirements as the single significant covariate. The non-compartmental analysis led to almost identical estimates of key parameters.

Conclusion: A high intra- and inter-patient variability was noted in the volume of distribution and clearance of rFVIIa in trauma patients with severe bleeding, mainly related with the transfusion requirements and thus blood loss and/or bleeding rate.

Figures

Figure 1
Figure 1
Factor VII (FVII) coagulant activity measurements in recombinant FVIIa-treated blunt (n = 6) and penetrating (n = 15) trauma patients. One outlier was excluded from the population pharmacokinetic model data set.
Figure 2
Figure 2
Correlation between clearance and red blood cell (RBC) transfusion requirement after first dose of recombinant factor VIIa (rFVIIa). Plot of model-estimated, individual clearance versus measured post-dose RBC requirement (n= 123). R-squared value reflects the fraction of the variation in individual predicted clearance values explained by the model. P value reflects the significance level at which the hypothesis of no effect of RBC transfusion need after rFVIIa dosing on individual clearance was rejected. Dotted lines indicate 95% confidence interval of the regression curve.
Figure 3
Figure 3
Population factor VII (FVII) coagulant activity profile modeled from the study dosing regimen. Dots represent the observed FVII coagulant activities from both frequent and sparse sampling while the solid line is trauma average population profile for multiple dosing. This model shows dosing in an adult hemophilia population superimposed for comparison (see Materials and methods).
Figure 4
Figure 4
Population pharmacokinetics profiles simulated at various red blood cell (RBC) transfusion requirements (20, 30, and 40 units after dosing) – increasing transfusion requirement linked with increasing clearance. The full line depicts the global trauma population (Trauma, mean of 8.7 RBC units) Data for a hemophilia population has been superimposed for comparison(see Materials and methods).

References

    1. Abshire T, Kenet G. Recombinant factor VIIa: review of efficacy, dosing regimens and safety in patients with congenital and acquired factor VIII or IX inhibitors. J Thromb Haemost. 2004;2:899–909. doi: 10.1111/j.1538-7836.2004.00759.x.
    1. Hoffman M, Monroe DM. A cell-based model of haemostasis. Thromb Haemost. 2001;85:958–965.
    1. Monroe DM, Hoffman M, Oliver JA, Roberts HR. Platelet activity of high dose factor VIIa is independent of tissue factor. Br J Haematol. 1997;99:542–547. doi: 10.1046/j.1365-2141.1997.4463256.x.
    1. Carr ME, Martin EJ. Recombinant factor VIIa: clinical applications for an intravenous haemostatic agent with broad-spectrum potential. Expert Rev Cardiovasc Ther. 2004;2:661–674. doi: 10.1586/14779072.2.5.661.
    1. Roberts HR, Monroe DM, Escobar MA. Current concepts of hemostasis: implications for therapy. Anesthesiology. 2004;100:885–893. doi: 10.1097/00000542-200404000-00019.
    1. Malherbe S, Tsui BC, Stobart K, Koller J. Argatroban as anticoagulant in cardiopulmonary bypass in an infant and attempted reversal with recombinant activated factor VII. Anesthesiology. 2004;101:1048–1049.
    1. Martinowitz U, Kenet G, Segal E, Luboshitz J, Lubetsky A, Ingerslev J, Lynn M. Recombinant activated factor VII for adjunctive hemorrhage control in trauma. J Trauma. 2001;51:431–439.
    1. Dutton RP, McCunn M, Hyder M, D'Angelo M, O'Connor JO, Hess JR, Scalea TM. Factor VIIa for correction of traumatic coagulopathy. J Trauma. 2004;57:709–719.
    1. Lodge JP, Jonas S, Oussoultzoglou E, Malago M, Jayr C, Cherqui D, Anthuber M, Mirza DF, Juhlman L, Bechstein WO, et al. Recombinant coagulation factor VIIa in major liver resection: a randomized, placebo-controlled, double blind clinical trial. Anesthesiology. 2005;102:269–275. doi: 10.1097/00000542-200502000-00006.
    1. Boffard KD, Riou B, Warren B, Iau P, Rizoli S, Kluger Y, Rossaint R, Axelsen M, The NovoSeven® Trauma Study Group Recombinant Factor VIIa as adjunctive therapy for bleeding control in severely injured trauma patients. A randomized, placebo-controlled, double-blind clinical trial. J Trauma. 2005;59:8–18.
    1. Allen GA, Monroe III, Roberts HR, Hoffman M. The effect of factor X level on thrombin generation and the procoagulant effect of activated factor VII in a cell-based model of coagulation. Blood Coagul Fibrinolysis. 2000;11(Suppl 1):S3–S7.
    1. Santagostino E, Morfini M, Rocino A, Baudo F, Scaraggi FA, Gringeri A. Relationship between factor VII activity and clinical efficacy of recombinant factor VIIa given by continuous infusion in patients with factor VIII inhibitors. Thromb Haemost. 2001;86:954–958.
    1. Villar A, Aronis S, Morfini M, Santagostino E, Auerswald G, Thomsen HF, Erhardsten E, Giangrande PC. Pharmacokinetics of activated recombinant coagulation factor VII (NovoSeven) in children vs adults with haemophilia A. Haemophilia. 2004;10:352–359. doi: 10.1111/j.1365-2516.2004.00925.x.
    1. Erhardtsen E. Pharmacokinetics of recombinant activated factor VII (rFVIIa) Semin Thromb Hemost. 2000;26:385–391. doi: 10.1055/s-2000-8457.
    1. Morrissey JH, Macik BG, Neunenschwander PF, Comp PC. Quantification of activated FVII levels in plasma using a tissue factor mutant selectively deficient in promoting factor VII activation. Blood. 1993;81:734–744.
    1. Aarons L. Population pharmacokinetics: Theory and practice. Br J Clin Pharmacol. 1991;32:669–670.
    1. Ette EI, Williams PJ. Population pharmacokinetics. I: Background, concepts, and models. Ann Pharmacother. 2004;38:1702–1706. doi: 10.1345/aph.1D374.
    1. Lindley CM, Sawyer WT, Macik BG, Lusher J, Harrison JF, Baird-Cox K, Birch K, Glazer S, Roberts HR. Pharmacokinetics and pharmacodynamics of recombinant factor VIIa. Clin Pharmacol Ther. 1994;55:638–648.
    1. The NONMEM Project Group . NONMEM Users Guide, November 1994, Part V: Introductory Guide. San Francisco; University of California;
    1. Fridberg MJ, Hedner U, Roberts HR, Erhardtsen E. A study of the pharmacokinetics and safety of recombinant activated factor VII in healthy Caucasian and Japanese subjects. Blood Coagul Fibrinolysis. 2005;16:259–266. doi: 10.1097/01.mbc.0000169218.15926.34.
    1. Girard P, Nony P, Erhardtsen E, Delair F, French P, Dechavanne M, Boissel JP. Population pharmacokinetics of recombinant factor VIIa in volunteers anticoagulated with acenocoumarol. Thromb Haemost. 1998;80:109–113.
    1. Kurita T, Kazama T, Morita K, Fujii S, Uraoka M, Takata K, Sato S. Influence of fluid infusion associated with high-volume blood loss on plasma propofol concentrations. Anesthesiology. 2004;100:871–878. doi: 10.1097/00000542-200404000-00017.
    1. Johnson KB, Egan TD, Kern SE, McJames SW, Cluff ML, Pace NL. Influence of hemorrhagic shock followed by crystalloid resuscitation on propofol: pharmacokinetic and pharmacodynamic analysis. Anesthesiology. 2004;101:647–659. doi: 10.1097/00000542-200409000-00013.
    1. Karkouti K, Beattie WS, Wijeysundera DN, Yau TM, McCluskey SA, Ghannam M, Sutton D, van Rensburg A, Karski J. Recombinant factor VIIa for intractable blood loss after cardiac surgery: a propensity score-matched case-control analysis. Transfusion. 2005;45:26–34. doi: 10.1111/j.1537-2995.2005.04216.x.
    1. Weiskopf RB. Intraoperative use of recombinant activated coagulation factor VII. Anesthesiology. 2002;96:1287–1289. doi: 10.1097/00000542-200206000-00004.
    1. Martinowitz U, Kenet G, Lubetski A, Luboshitz J, Segal E. Possible role of recombinant activated factor VII (rFVIIa) in the control of hemorrhage associated with massive trauma. Can J Anaesth. 2002;49(Suppl):S15–S20.
    1. Barletta JF, Ahrens CL, Tyburski JG, Wilson RF. A review of recombinant factor VII for refractory bleeding in non-hemophilic trauma patients. J Trauma. 2005;58:646–651.
    1. Kurita T, Kazama T, Morita K, Fujii S, Uraoka M, Takata K, Sato S. Influence of fluid infusion associated with high-volume blood loss on plasma propofol concentrations. Anesthesiology. 2004;100:871–878. doi: 10.1097/00000542-200404000-00017.

Source: PubMed

3
Tilaa