Pharmacokinetics of plasma-derived C1-esterase inhibitor after subcutaneous versus intravenous administration in subjects with mild or moderate hereditary angioedema: the PASSION study

Inmaculada Martinez-Saguer, Marco Cicardi, Chiara Suffritti, Eva Rusicke, Emel Aygören-Pürsün, Hildegard Stoll, Tanja Rossmanith, Annette Feussner, Uwe Kalina, Wolfhart Kreuz, Inmaculada Martinez-Saguer, Marco Cicardi, Chiara Suffritti, Eva Rusicke, Emel Aygören-Pürsün, Hildegard Stoll, Tanja Rossmanith, Annette Feussner, Uwe Kalina, Wolfhart Kreuz

Abstract

Background: Hereditary angioedema (HAE) is a rare disease caused by C1-esterase inhibitor (C1-INH) deficiency, characterized by periodic attacks of acute edema affecting subcutaneous (SC) tissues and mucous membranes. Human C1-INH concentrate given intravenously (IV) is effective and safe, but venous access may be difficult. We compared SC and IV administration of human pasteurized C1-INH concentrate with respect to pharmacokinetics, pharmacodynamics, and safety.

Study design and methods: This was a prospective, randomized, open-label, crossover study. Twenty-four subjects with mild or moderate HAE were randomly assigned during an attack-free interval to receive 1000 units of human pasteurized C1-INH concentrate IV or SC. Plasma levels of C1-INH activity and antigen, C4 antigen, cleaved high-molecular-weight kininogen (clHK), and C1-INH antibodies were measured.

Results: The mean relative bioavailability of functional C1-INH after SC administration was 39.7%. Maximum C1-INH activity after SC administration occurred within 48 hours and persisted longer than after IV administration. C4 antigen levels increased and clHK levels decreased after IV and SC administration, indicating the pharmacodynamic action of C1-INH. The mean half-life of functional C1-INH was 62 hours after IV administration and 120 hours after SC administration (p=0.0595). C1-INH concentrate was safe and well tolerated when administered via both routes. As expected, SC administration resulted in a higher incidence of injection site reactions, all of which were mild.

Conclusion: With a relative bioavailability of 39.7%, SC administration of human pasteurized C1-INH yields potentially clinically relevant and sustained plasma levels of C1-INH and is safe and well tolerated.

Trial registration: ClinicalTrials.gov NCT00748202.

© 2013 The Authors. Transfusion published by Wiley Periodicals, Inc. on behalf of AABB.

Figures

Figure 1
Figure 1
Mean C1-INH activity over time (pharmacokinetic per-protocol set, n = 23 subjects). Black and gray whiskers show SDs. Median values are indicated with “X.” Horizontal lines show baseline (BL) values (before first administration of study drug). Data for mean C1-INH activity at 336 and 504 hours were obtained in only six subjects. () IV; () SC.
Figure 2
Figure 2
Mean concentrations of C1-INH antigen over time (pharmacokinetic per-protocol set, n = 16 subjects). Seven subjects with HAE Type II were excluded from the C1-INH antigen analyses. Black and gray whiskers show SDs. Median values are indicated with “X.” Horizontal lines show baseline (BL) values (before first administration of study drug). Data for mean concentrations of C1-INH antigen at 336 and 504 hours were obtained in only six subjects. () IV; () SC.
Figure 3
Figure 3
Mean concentrations of C4 antigen over time (pharmacokinetic per-protocol set, n = 23 subjects). Black and gray whiskers show SDs. Median values are indicated with “X.” Horizontal lines show baseline (BL) values (before first administration of study drug). Data for mean concentrations of C4 antigen at 336 and 504 hours were obtained in only six subjects. () IV; () SC.
Figure 4
Figure 4
Mean clHK over time (pharmacokinetic per-protocol set, n = 6 subjects). Black and gray whiskers show SDs. Median values are indicated with “X.” Horizontal lines show baseline values (before first administration of study drug). () IV; () SC.

References

    1. Donaldson VH, Evans RR. A biochemical abnormality in hereditary angioneurotic oedema: absence of serum inhibitor of C1-esterase. Am J Med. 1963;35:37–44.
    1. Bork K, Siedlecki K, Bosch S, et al. Asphyxiation by laryngeal edema in patients with hereditary angioedema. Mayo Clin Proc. 2000;75:349–354.
    1. Frank MM, Gelfand JA, Atkinson JP. Hereditary angioedema: the clinical syndrome and its management. Ann Intern Med. 1976;84:580–593.
    1. Longhurst H, Cicardi M. Hereditary angio-edema. Lancet. 2012;379:474–481.
    1. Wuillemin WA, Hack CE, Bleeker WK, et al. Inactivation of factor XIa in vivo: studies in chimpanzees and in humans. Thromb Haemost. 1996;76:549–555.
    1. Garcia JG, Siflinger-Birnboim A, Bizios R, et al. Thrombin-induced increase in albumin permeability across the endothelium. J Cell Physiol. 1986;128:96–104.
    1. Garcia JG, Pavalko FM, Patterson CE. Vascular endothelial cell activation and permeability responses to thrombin. Blood Coagul Fibrinolysis. 1995;6:609–626.
    1. Cugno M, Cicardi M, Bottasso B, et al. Activation of the coagulation cascade in C1-inhibitor deficiencies. Blood. 1997;89:3213–3218.
    1. Bork K. Diagnosis and treatment of hereditary angioedema with normal C1 inhibitor. Allergy Asthma Clin Immunol. 2010;6:15.
    1. Hack CE. 2004. Pathogenesis of angioedema attacks. In: Hereditary and acquired angioedema: problems and progress. Proceedings of the third C1 esterase inhibitor deficiency workshop and beyond. JACI 114:S75–9.
    1. Craig TJ, Levy RJ, Wasserman RL, et al. Efficacy of human C1 esterase inhibitor concentrate compared with placebo in acute hereditary angioedema attacks. J Allergy Clin Immunol. 2009;124:801–808.
    1. Craig TJ, Wasserman RL, Levy RJ, et al. Prospective study of rapid relief provided by C1 esterase inhibitor in emergency treatment of acute laryngeal attacks in hereditary angioedema. J Clin Immunol. 2010;30:823–829.
    1. Wasserman RL, Levy RJ, Bewtra AK, et al. Prospective study of C1 esterase inhibitor in the treatment of successive acute abdominal and facial hereditary angioedema attacks. Ann Allergy Asthma Immunol. 2011;106:62–68.
    1. Bork K. Human pasteurized C1-inhibitor concentrate for the treatment of hereditary angioedema due to C1-inhibitor deficiency. Expert Rev Clin Immunol. 2011;7:723–733.
    1. Bork K, Korger G, Kreuz W. Review of the long-term safety of a human pasteurized C1 inhibitor concentrate. J Allergy Clin Immunol. 2012;129:AB222. Poster presented at the 2012 AAAAI Annual Meeting, Orlando, Florida.
    1. Bowen T, Cicardi M, Farkas H, et al. International consensus algorithm for the diagnosis, therapy and management of hereditary angioedema. Allergy Asthma Clin Immunol. 2010;6:24.
    1. Berrettini M, Lämmle B, White T, et al. Detection of in vitro and in vivo cleavage of high molecular weight kininogen in human plasma by immunoblotting with monoclonal antibodies. Blood. 1986;68:455–462.
    1. Craig T, Pürsün EA, Bork K, et al. WAO Guideline for the management of hereditary angioedema. World Allergy Organ J. 2012;5:182–199.
    1. Jiang H, Zhang HM, Frank MM. Subcutaneous infusion of human C1 inhibitor in swine. Clin Immunol. 2010;136:323–328.
    1. Schranz J, Levy R, Lumry W, et al. Safety, pharmacokinetics (PK), and pharmacodynamics (PD) of subcutaneous (SC) Cinryze® C1 inhibitor (C1 INH) with recombinant human hyaluronidase (rHuPH20) in subjects with hereditary angioedema (HAE) J Allergy Clin Immunol. 2012;129:AB369. Poster presented at the 2012 AAAAI Annual Meeting, Orlando, Florida.
    1. . 2012. A study to evaluate the safety, pharmacokinetics, and pharmacodynamics of subcutaneous CINRYZE administration. [cited 2013 May 13]. Accessed from: .
    1. Zuraw BL, Sugimoto S, Curd JG. The value of rocket immunoelectrophoresis for C4 activation in the evaluation of patients with angioedema or C1-inhibitor deficiency. J Allergy Clin Immunol. 1986;78:1115–1120.
    1. Davis AE., 3rd Hereditary angioedema: a current state-of-the-art review, III: mechanisms of hereditary angioedema. Ann Allergy Asthma Immunol. 2008;100:S7–12.
    1. Lämmle B, Zuraw BL, Heeb MJ, et al. Detection and quantitation of cleaved and uncleaved high molecular weight kininogen in plasma by ligand blotting with radiolabeled plasma prekallikrein or factor IX. Thromb Haemost. 1988;59:151–161.
    1. Nielsen EW, Johansen HT, Hogasen K, et al. Activation of the complement, coagulation, fibrinolytic and kallikrein-kinin systems during attacks of hereditary angioedema. Scand J Immunol. 1996;44:185–192.
    1. Cugno M, Cicardi M, Coppola R, et al. Activation of factor XII and cleavage of high molecular weight kininogen during acute attacks in hereditary and acquired C1-inhibitor deficiencies. Immunopharmacology. 1996;33:361–364.
    1. Bühler R, Hovinga JK, Aebi-Huber I, et al. Improved detection of proteolytically cleaved high molecular weight kininogen by immunoblotting using an antiserum against its reduced 47 kDa light chain. Blood Coagul Fibrinolysis. 1995;6:223–232.
    1. McDonald TC. Subcutaneous administration of biotherapeutics. Curr Opin Mol Ther. 2010;12:461–470.
    1. Späth PJ, Wüthrich B, Bütler R. Quantification of C1-inhibitor functional activities by immunodiffusion assay in plasma of patients with hereditary angioedema–evidence of a functionally critical level of C1-inhibitor concentration. Complement. 1984;1:147–159.
    1. Bork K, Witzke G. Long-term prophylaxis with C1-inhibitor (C1 INH) concentrate in patients with recurrent angioedema caused by hereditary and acquired C1-inhibitor deficiency. J Allergy Clin Immunol. 1989;83:677–682.
    1. Farkas H, Harmat G, Füst G, et al. Clinical management of hereditary angio-oedema in children. Pediatr Allergy Immunol. 13:153–161.
    1. Levi M, Choi G, Picavet C, et al. Self-administration of C1-inhibitor concentrate in patients with hereditary or acquired angioedema caused by C1-inhibitor deficiency. J Allergy Clin Immunol. 2006;117:904–908.
    1. Kaplan AP, Joseph K. The bradykinin-forming cascade and its role in hereditary angioedema. Ann Allergy Asthma Immunol. 2010;104:193–204.
    1. Bernstein JA, Ritchie B, Levy RJ, et al. Population pharmacokinetics of plasma-derived C1 esterase inhibitor concentrate used to treat acute hereditary angioedema attacks. Ann Allergy Asthma Immunol. 2010;105:149–154.
    1. Martinez-Saguer I, Rusicke E, Aygören-Pürsün E, et al. Pharmacokinetic analysis of human plasma-derived pasteurized factor concentrate in adults and children with hereditary angioedema: a prospective study. Transfusion. 2010;50:354–360.
    1. European Medicines Agency. Cinryze—assessment report. Procedure No. EMEA/H/C/001207. [cited 2013 Nov 13]. Available from: .
    1. European Medicines Agency. Cinryze 500 Units powder and solvent for solution for injection. Summary of product characteristics. [cited 2013 Nov 13]. Available from: .
    1. Brackertz D, Isler E, Kueppers F. Half-life of C1-INH in hereditary angioneurotic oedema (HAE) Clin Allergy. 1975;1:89–94.
    1. Skoda-Smith S, Torgerson TR, Ochs HD. Subcutaneous immunoglobulin replacement therapy in the treatment of patients with primary immunodeficiency disease. Ther Clin Risk Manag. 2010;6:1–10.
    1. Craig TJ, Bewtra AK, Bahna SL, et al. C1 esterase inhibitor concentrate in 1085 hereditary angioedema attacks—final results of the I.M.P.A.C.T.2 study. Allergy. 2011;66:1604–1611.
    1. Braun A, Kwee L, Labow MA, et al. Protein aggregates seem to play a key role among the parameters influencing the antigenicity of interferon alpha (IFN-alpha) in normal and transgenic mice. Pharm Res. 1997;14:1472–1478.
    1. Kromminga A, Schellekens H. Antibodies against erythropoietin and other protein-based therapeutics: an overview. Ann N Y Acad Sci. 2005;1050:257–265.
    1. Schellekens H, Jiskoot W. Erythropoietin-associated PRCA: still an unsolved mystery. J Immunotoxicol. 2006;3:123–130.

Source: PubMed

3
Předplatit