Favorable pharmacokinetics in hemophilia B for nonacog beta pegol versus recombinant factor IX-Fc fusion protein: A randomized trial

Carmen Escuriola Ettingshausen, Inga Hegemann, Mindy L Simpson, Adam Cuker, Roshni Kulkarni, Rajiv K Pruthi, May-Lill Garly, Rikke M Meldgaard, Paula Persson, Robert Klamroth, Carmen Escuriola Ettingshausen, Inga Hegemann, Mindy L Simpson, Adam Cuker, Roshni Kulkarni, Rajiv K Pruthi, May-Lill Garly, Rikke M Meldgaard, Paula Persson, Robert Klamroth

Abstract

Background and objective: Nonacog beta pegol (N9-GP) and recombinant factor IX-Fc fusion protein (rFIXFc) are extended half-life rFIX compounds. We report the first single-dose pharmacokinetic trial of N9-GP and rFIXFc.

Patients/methods: Paradigm 7 was a multicenter, open-label, randomized, crossover trial in previously treated (>150 exposure days) adults with congenital hemophilia B (FIX activity ≤2%). Patients received single intravenous injections (50 IU/kg) of N9-GP and rFIXFc with at least 21 days between doses. Plasma FIX activity, predose, and at serial time points up to 240 hours postdose, was measured using validated one-stage clotting assays (SynthAFax for N9-GP; Actin FSL for rFIXFc) and a chromogenic assay (ROX factor IX) with normal human plasma as calibrator. The primary endpoint was area under the FIX activity-time curve from 0 to infinity, dose-normalized to 50 IU/kg (AUC0-inf,norm).

Results: Fifteen patients received study treatment. Based on FIX activity results from the one-stage clotting assays, estimated AUC0-inf,norm was significantly greater for N9-GP than rFIXFc (ratio: 4.39; P <0.0001, based on a two-sided test on 5% significance level). In addition, N9-GP had a longer terminal half-life, two times higher incremental recovery at 30 minutes and maximum FIX activity (dose-normalized to 50 IU/kg) and six times higher FIX activity at 168 hours than rFIXFc. These findings were largely comparable with the chromogenic assay data and are consistent with published data for each compound.

Conclusions: In this comparison, N9-GP demonstrated favorable pharmacokinetic characteristics versus rFIXFc, helping clinicians to understand differences between N9-GP and rFIXFc.

Registration: This trial is registered with clinicaltrials.gov (NCT03075670) and the European Clinical Trials Database (EudraCT: 2016-001149-25).

Keywords: crossover trial; factor IX; hemophilia B; pharmacokinetics; phase I; randomized clinical trial.

Figures

Figure 1
Figure 1
Trial design and pharmacokinetic sampling. Doses of each trial product were separated by at least 21 d; each dose administration was preceded by a 96‐h washout from non‐modified FIX products. FIX, factor IX; PK, pharmacokinetic; rFIXFc, recombinant factor IX‐Fc fusion protein; V, visit
Figure 2
Figure 2
Mean FIX activity profiles following single 50‐IU/kg doses of N9‐GP and rFIXFc, measured using one‐stage clotting and chromogenic assays. Mean (±SD) FIX activity profiles following single 50‐IU/kg doses of N9‐GP and rFIXFc are shown, measured using one‐stage clotting assays (A) and the chromogenic assay (B). Panel (C) collates the mean plots with an insert magnifying FIX activity over the first 24 h. The aPTT reagents used for the one‐stage clotting assay were SynthAFax and Actin FSL for N9‐GP and rFIXFc, respectively. The ROX factor IX kit was used for the chromogenic assay with both treatments. All assays were calibrated using NHP. aPTT, activated partial thromboplastin time; FIX, factor IX; NHP, normal human plasma; PK, pharmacokinetic, rFIXFc, recombinant factor IX‐Fc fusion protein; SD, standard deviation
Figure 3
Figure 3
Analysis of PK endpoints for N9‐GP versus rFIXFc, derived from FIX activity measured using one‐stage clotting and chromogenic assays. All PK endpoints were log‐transformed before being analyzed using a mixed‐effects model that included product and period as fixed effects and patients as a random effect. Estimates with two‐sided 95% CIs were provided for each treatment, back‐transformed to the original scale; the two‐sided 95% CIs for the comparisons between treatments were expressed as ratios and provided together with the P‐values. The aPTT reagents used for the one‐stage clotting assay were SynthAFax and Actin FSL for N9‐GP and rFIXFc, respectively. The ROX factor IX kit was used for the chromogenic assay with both treatments. All assays were calibrated using NHP. *< 0.0001 for all comparisons, except for t½, for which P < 0.001 for both assays. aPTT, activated partial thromboplastin time; AUC0‐inf,norm, area under the FIX activity–time curve from 0 to infinity, dose‐normalized to 50 IU/kg; C168h, FIX activity at 168 h; C240h, FIX activity at 240 h; CI, confidence interval; CL, clearance; Cmax,norm, maximum FIX activity dose‐normalized to 50 IU/kg; FIX, factor IX; IR30min, incremental recovery at 30 min; NHP, normal human plasma; PK, pharmacokinetic; rFIXFc, recombinant factor IX‐Fc fusion protein; t½, terminal half‐life; Vss, apparent volume of distribution at steady state
Figure 4
Figure 4
Log‐scale mean FIX activity profiles following single 50‐IU/kg doses of N9‐GP and rFIXFc, measured using one‐stage clotting and chromogenic assays. Log‐scale mean FIX activity profiles following single 50‐IU/kg doses of N9‐GP and rFIXFc are shown, measured using one‐stage clotting assays and the chromogenic assay. The aPTT reagents used for the one‐stage clotting assay were SynthAFax and Actin FSL for N9‐GP and rFIXFc, respectively. The ROX factor IX kit was used for the chromogenic assay with both treatments. All assays were calibrated using NHP. aPTT, activated partial thromboplastin time; FIX, factor IX; NHP, normal human plasma; rFIXFc, recombinant factor IX‐Fc fusion protein

References

    1. Carcao M. Changing paradigm of prophylaxis with longer acting factor concentrates. Haemophilia. 2014;20:99–105.
    1. Manco‐Johnson MJ, Abshire TC, Shapiro AD, Riske B, Hacker MR, Kilcoyne R, et al. Prophylaxis versus episodic treatment to prevent joint disease in boys with severe hemophilia. N Engl J Med. 2007;357:535–44.
    1. Gringeri A, Lundin B, von Mackensen S, Mantovani L, Mannucci PM; ESPRIT Study Group . A randomized clinical trial of prophylaxis in children with hemophilia A (the ESPRIT Study). J Thromb Haemost. 2011;9:700–10.
    1. Iorio A, Marchesini E, Marcucci M, Stobart K, Chan AK. Clotting factor concentrates given to prevent bleeding and bleeding‐related complications in people with hemophilia A or B. Cochrane Database Syst Rev. 2011:CD003429.
    1. Iorio A, Blanchette V, Blatny J, Collins P, Fischer K, Neufeld E. Estimating and interpreting the pharmacokinetic profiles of individual patients with hemophilia A or B using a population pharmacokinetic approach: communication from the SSC of the ISTH. J Thromb Haemost. 2017;15:2461–5.
    1. Collins PW, Fischer K, Morfini M, Blanchette VS, Bjorkman S; International Prophylaxis Study Group Pharmacokinetics Expert Working Group . Implications of coagulation factor VIII and IX pharmacokinetics in the prophylactic treatment of haemophilia. Haemophilia. 2011;17:2–10.
    1. Lissitchkov T, Matysiak M, Zavilska K, Laguna P, Gercheva L, Antonov A, et al. Head‐to‐head comparison of the pharmacokinetic profiles of a high‐purity factor IX concentrate (AlphaNine®) and a recombinant factor IX (BeneFIX®) in patients with severe haemophilia B. Haemophilia. 2013;19:674–8.
    1. Refixia . [summary of product characteristics], Novo Nordisk. Bagsværd, Denmark. Available from: . Accessed March 6, 2018.
    1. Alprolix . [summary of product characteristics], Swedish Orphan Biovitrum Ltd. Stockholm, Sweden. Available from: . Accessed March 6, 2018.
    1. Negrier C, Knobe K, Tiede A, Giangrande P, Moss J. Enhanced pharmacokinetic properties of a glycoPEGylated recombinant factor IX: a first human dose trial in patients with hemophilia B. Blood. 2011;118:2695–701.
    1. Collins PW, Young G, Knobe K, Karim FA, Angchaisuksiri P, Banner C, et al.; paradigm 2 Investigators . Recombinant long‐acting glycoPEGylated factor IX in hemophilia B: a multinational randomized phase 3 trial. Blood. 2014;124:3880–6.
    1. Young G, Collins PW, Colberg T, Chuansumrit A, Hanabusa H, Lentz SR, et al. Nonacog beta pegol (N9‐GP) in haemophilia B: a multinational phase III safety and efficacy extension trial (paradigm4). Thromb Res. 2016;141:69–76.
    1. Carcao M, Zak M, Abdul Karim F, Hanabusa H, Kearney S, Lu MY, et al. Nonacog beta pegol in previously treated children with hemophilia B: results from an international open‐label phase 3 trial. J Thromb Haemost. 2016;14:1521–276.
    1. Shapiro AD, Ragni MV, Valentino LA, Key NS, Josephson NC, Powell JS, et al. Recombinant factor IX‐Fc fusion protein (rFIXFc) demonstrates safety and prolonged activity in a phase 1/2a study in hemophilia B patients. Blood. 2012;119:666–72.
    1. Powell JS, Pasi KJ, Ragni MV, Ozelo MC, Valentino LA, Mahlangu JN, et al. Phase 3 study of recombinant factor IX Fc fusion protein in hemophilia B. N Engl J Med. 2013;369:2313–23.
    1. Powell JS, Apte S, Chambost H, Hermans C, Jackson S, Josephson NC, et al. Long‐acting recombinant factor IX Fc fusion protein (rFIXFc) for perioperative management of subjects with haemophilia B in the phase 3 B‐LONG study. Br J Haematol. 2015;168:124–34.
    1. Fischer K, Kulkarni R, Nolan B, Mahlangu J, Rangarajan S, Gambino G, et al. Recombinant factor IX Fc fusion protein in children with haemophilia B (Kids B‐LONG): results from a multicentre, non‐randomised phase 3 study. Lancet Haematol. 2017;4:e75–82.
    1. Tiede A, Abdul‐Karim F, Carcao M, Persson P, Clausen WHO, Kearney S, et al. Pharmacokinetics of a novel extended half‐life glycoPEGylated factor IX, nonacog beta pegol (N9‐GP) in previously treated patients with haemophilia B: results from two phase 3 clinical trials. Haemophilia. 2017;23:547–55.
    1. Iorio A, Edginton AN, Blanchette V, Blatny J, Boban A, Cnossen M, et al. Performing and interpreting individual pharmacokinetic profiles in patients with Hemophilia A or B: rationale and general considerations. Res Pract Thromb Haemost. 2018;2:535–48.
    1. International Conference on Harmonisation ICH harmonised tripartite guideline . Guideline for good clinical practice E6(R1). Current Step 4 version. 1996. Available from: . Accessed March 6, 2018.
    1. World Medical Association . WMA Declaration of Helsinki ‐ ethical principles for medical research involving human subjects. Last amended by the 64th WMA General Assembly, Fortaleza, Brazil. Available at: . Accessed March 6, 2018.
    1. Sommer JM, Buyue Y, Bardan S, Peters RT, Jiang H, Kamphaus GD, et al. Comparative field study: impact of laboratory assay variability on the assessment of recombinant factor IX Fc fusion protein (rFIXFc) activity. Thromb Haemost. 2014;112:932–40.
    1. Kershaw GW, Dissanayake K, Chen VM, Khoo TL. Evaluation of chromogenic factor IX assays by automated protocols. Haemophilia. 2018;24:492–501.
    1. Tiefenbacher S, Bohra R, Amiral J, Bowyer A, Kitchen S, Lochu A, et al. Qualification of a select one‐stage activated partial thromboplastin time‐based clotting assay and two chromogenic assays for the post‐administration monitoring of nonacog beta pegol. J Thromb Haemost. 2017;15:1901–12.
    1. Dargaud Y, Delavenne X, Hart DP, Meunier S, Mismetti P. Individualized PK‐based prophylaxis in severe haemophilia. Haemophilia. 2018;24:3–17.
    1. Fischer K, Ljung R. Primary prophylaxis in haemophilia care: guideline update 2016. Blood Cells Mol Dis. 2017;67:81–5.
    1. Diao L, Li S, Ludden T, Gobburu J, Nestorov I, Jiang H. Population pharmacokinetic modelling of recombinant factor IX Fc fusion protein (rFIXFc) in patients with haemophilia B. Clin Pharmacokinet. 2014;53:467–77.
    1. Feng D, Stafford KA, Broze GJ, Stafford DW. Evidence of clinically significant extravascular stores of factor IX. J Thromb Haemost. 2013;11:2176–8.
    1. Sun J, Hua B, Livingston EW, Taves S, Johansen PB, Hoffman M, et al. Abnormal joint and bone wound healing in hemophilia mice is improved by extending factor IX activity after hemarthrosis. Blood. 2017;129:2161–71.

Source: PubMed

3
S'abonner