Thromb Haemost 1993; 70(03): 458-464
DOI: 10.1055/s-0038-1649605
Original Article
Coagulation
Schattauer GmbH Stuttgart

Pharmacokinetics of Recombinant Factor VIIa in the Rat – A Comparison of Bio-, Immuno- and Isotope Assays

Mads Krogsgaard Thomsen
1   The Biopharmaceuticals Division, Research and Development, Novo Nordisk A/S, Gentofte, Denmark
,
Viggo Diness
1   The Biopharmaceuticals Division, Research and Development, Novo Nordisk A/S, Gentofte, Denmark
,
Povl Nilsson
1   The Biopharmaceuticals Division, Research and Development, Novo Nordisk A/S, Gentofte, Denmark
,
Søren Nørgaard Rasmussen
2   The Institute of Biology, Royal Danish School of Pharmacy, Copenhagen, Denmark
,
Trevor Taylor
3   The Department of Metabolism and Pharmacokinetics, Huntingdon Research Centre, Cambridgeshire, England
,
Ulla Hedner
1   The Biopharmaceuticals Division, Research and Development, Novo Nordisk A/S, Gentofte, Denmark
› Author Affiliations
Further Information

Publication History

Received 10 December 1992

Accepted after revision 06 May 1993

Publication Date:
05 July 2018 (online)

Summary

Recombinant human factor VII a (rFVIIa) is an activated coagulation factor for intravenous use as a haemostatic agent in haemophiliacs who generate antibodies against factor VIII or IX. Plasma kinetic studies are important for the understanding of the action of rFVIIa which is exerted in the vascular compartment of the body, more specifically on the vessel walls at the site of injury. In the present study, rats were dosed 100 or 500 μg/kg 125I-rFVIIa i. V., without any side effects being observed, and the plasma profile of rFVIIa was studied by 3 different assays that were shown to correlate well at early times post-dose: trichloroacetic acid (TCA)-precipitable drug-related radioactivity, rFVIIa antigen determination by ELISA technique, and the assay of clot activity which is the only clinically applicable assay. The plasma concentration curve could be resolved into 1-3 exponentials, depending on the FVIIa detection principle that was employed. Initially, there was a short (ca. 10 min) phase of increasing concentrations before the attainment of C max. This was followed by a plasma recovery (C max × plasma volume/dose) in the vicinity of one half of the administered dose. The initial volume of distribution (V 1) corresponded to the vascular compartment whereas the volume of distribution at steady state (V ss) was somewhat larger. Whole body clearance (CL-B) of rFVIIa was approx. 1 ml/min per kg, and mean residence time (MRT) and the half-life assumed to be associated with the loss of biological activity was approx. 1 h and 20-45 min, respectively. From these plasma data, rFVIIa appears to be a low clearance compound with limited tissue distribution and a short half-life. Tissue distribution studies showed that high 125I levels, assumed to be rFVIIa-related, included mineralised bone and well-perfused organs such as the liver which suggested that this organ was responsible for a major proportion of CL-B. Finally, mass balance studies showed that almost 90% of the administered radioactivity could be accounted for following an i. v. dose, predominantly as non drug-related radioactivity, even though a small amount of TCA-precipitable radioactivity was excreted via the biliary route. In conclusion, dose- or sex-dependent plasma kinetics and tissue distribution within a dose range of 100 to 500 μg/kg of rFVIIa was not observed. In the early and pharmacologically relevant phase after rFVIIa administration there appears to be good agreement between the various plasma assays employed in the study, indicating that the clot assay yields useful information in studies of rFVIIa plasma pharmacokinetics.

 
  • References

  • 1 Shapiro SS, Hultin M. Acquired inhibitors to the blood coagulation factors. Semin Thromb Hemostas 1975; 1: 336-385
  • 2 Fair DS. Quantitation of factor VII in the plasma of normal and warfarin-treated individuals by radioimmunoassay. Blood 1983; 62: 784-791
  • 3 Hedner U. Factor VIIa in the treatment of haemophilia. Blood Coag Fibrinol 1990; 1: 307-317
  • 4 Wildgoose PW, Jørgensen T, Komiyama Y, Nakagaki T, Pedersen A, Kiesel W. The role of phospholipids and the factor FVII Gla-domain in the interaction of factor VII with tissue factor. Thromb Haemostas 1992; 67: 679-685
  • 5 Babior BM, Stossel TP. Hematology. A Pathophysiological Approach. 2. Churchill Livingstone New York: 1984
  • 6 Hedner U, Kiesel W. Use of human factor VIIa in the treatment of two haemophilia A patients with high-titer inhibitors. J Clin Invest 1983; 71: 1836-1841
  • 7 Thim L, Bjørn S, Christensen M, Nicolaisen EM, Lund-Hansen T, Pedersen AH, Hedner U. Amino acid sequence and posttranslational modifications of human factor VII a from plasma and transfected baby hamster kidney cells. Biochemistry 1988; 27: 7785-7793
  • 8 Hedner U, Ljungberg J, Lund-Hansen T. Comparison of the effect of plasma-derived and recombinant human FVIIa in vitro and in a rabbit model. Blood Coag Fibrinol 1990; 1: 145-151
  • 9 Diness V, Bregengaard C, Erhardtsen E, Hedner U. Recombinant human factor VII a (rFVIIa) in a rabbit stasis model. Thromb Res 1992; 67: 233-241
  • 10 Brinkhous KM, Hedner U, Garris JB, Diness V, Read MS. Effect of recombinant factor VIIa on the haemostatic defect in dogs with haemophilia A, haemophilia B, and von Willebrand disease. Proc Natl Acad Sci 1989; 86: 1382-1386
  • 11 Diness V, Lund-Hansen T, Hedner U. Effect of recombinant human FVIIa on warfarin-induced bleeding in rats. Thromb Res 1990; 59: 921-929
  • 12 Pedersen AH, Lund-Hansen T, Bisgård-Franzen H, Olsen F, Petersen LC. Autoactivation of human recombinant coagulation factor VII. Biochemistry 1989; 28: 9331-9336
  • 13 Regoeczi E. Iodine-labelled plasma protein, Vol. I. CRC Press; Boca Raton, FL: 1984
  • 14 Lund-Hansen T, Olsen AA, Selmer J. A double monoclonal ELISA for human coagulation factor VII. Thromb Haemostas 1989; 62: 55 (abstract)
  • 15 Broze G, Majerus PW. Human factor VII. Methods Enzymol 1981; 10: 228-237
  • 16 Olsson DM. A sequential Simplex program for solving minimization Problems. J Quality Tech 1974; 6: 53-57
  • 17 Yamaoka K, Tanigawara Y, Nakagawa T, Uno T. A pharmacokinetic analysis program (multi) for microcomputer. J Pharm Dyn 1981; 4: 879-885
  • 18 Morfini M, Lee M, Messori A. The design and analysis of half-life and recovery studies for factor VII and factor IX. Thromb Haemostas 1991; 66: 384-386
  • 19 Baker HJ, Lindsey JR, Weisbroth SH. Selected normative data. In: The Laboratory Rat, Vol. II. Academic Press; New York: 1980
  • 20 Hedner U, Glazer S, Pingel K, Alberts KA, Blombäck M, Schulman S, Johnson H. Successful use of recombinant Factor VII a in a patient with severe hemophilia A during synovectomy. Lancet 1988; II: 1193
  • 21 Wildgoose P, Nemerson Y, Hansen LL, Nielsen FE, Glazer S, Hedner U. Measurement of basal levels of factor VII a in Haemophilia A and B patients. Blood 1992; 80: 25-28
  • 22 Mannucci PM, Morfini M. Determination of pharmacokinetics of replacement factor VIII. Semin Hematol 1990; 27 (Suppl. 02) 8-10
  • 23 Schwarz RS, Abildgaard CF, Aledort LM, Arkin S, Bloom AL, Brackmann HH, Brettler DB, Fukui H, Hilgartner MW, Inwood MJ, Kasper CK, Kernof PBA, Levine PH, Lusher JM, Mannucci PM, Scharrer I, MacKenzie MA, Pancham N, Kuo HS, Allred RU. Human recombinant DNA-derived antihemophilic factor (factor VIII) in the treatment of hemophilia A. N Engl J Med 1990; 323: 1800-1805
  • 24 Flamenbaum W, Huddleston ML, McNeil JS, Hamburger RJ. Uranyl nitrate-induced acute renal failure in the rat: Micropuncture and renal hemodynamic studies. Kidney Int 1974; 6: 408-418
  • 25 Østerud B, Miller-Anderson M, Abilgaard U, Pryz H. The effect of anti-thrombin III on the activity of the coagulation factors VII, IX and X. Thromb Haemostas 1976; 35: 295-304
  • 26 Vainieri H, Wingard LB. Effect of warfarin on the kinetics of the vitamin K-dependent clotting factors in the rat. J Pharmacol Exp Therap 1977; 201: 507-517
  • 27 Seligsohn U, Kasper CK, Østerud B, Rapaport SI. Activated factor VII: Presence in factor IX concentrates and persistence in the circulation after infusion. Blood 1979; 53: 828-837
  • 28 Hasselback R, Hjort PF. Effect of heparin on in vivo turnover of clotting factors. J Appl Physiol 1960; 15: 945-948
  • 29 Loeliger EA, Esch BVD, Cleton FJ, Booij HL, Mattern MJ. On the metabolism of factor VII. In: Proc. 7th Congr Eur Soc. Haematol SKarger. Basel: 1960: 160
  • 30 Thomsen MK, Wildgoose P, Nilsson P, Hedner U. Accumulation of recombinant factor VIIa in rat bone: Importance of the Gla-domain and relevance to factor IX, another vitamin K-dependent clotting factor. Pharmacol Toxicol. In press
  • 31 Beeby T, Chasseaud LF, Taylor T, Thomsen MK. Distribution of the recombinant coagulation factor 125 I-rFVIIa in rats. Thromb Haemostas 1993; 70: 465-468
  • 32 Furie JS, Furie BC. Molecular and cellular biology of blood coagulation. N Engl J Med 1992; 326: 800-806