Thromb Haemost 2020; 120(06): 968-976
DOI: 10.1055/s-0040-1710315
Coagulation and Fibrinolysis
Georg Thieme Verlag KG Stuttgart · New York

Emicizumab Improves Ex Vivo Clotting Function in Patients with Mild/Moderate Hemophilia A

Yuto Nakajima
1   Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
,
Keiji Nogami
1   Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
,
Koji Yada
1   Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
2   The Course of Hemophilia Education, Nara Medical University, Nara, Japan
,
Shoko Furukawa
1   Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
3   The Course of Thrombosis and Hemostasis Molecular Pathology, Nara Medical University, Nara, Japan
,
Mariko Noguchi-Sasaki
4   Chugai Pharmaceutical Co., Ltd, Division of Product Research, Kamakura, Kanagawa, Japan
,
Michinori Hirata
4   Chugai Pharmaceutical Co., Ltd, Division of Product Research, Kamakura, Kanagawa, Japan
,
Midori Shima
1   Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
› Author Affiliations
Funding This work was partly supported by a Grant-in-Aid for Scientific Research (KAKENHI) from the Ministry of Education, Culture, Sports, Science and Technology (MEXT) to K.N. (18K07885).
Further Information

Publication History

20 January 2020

01 April 2020

Publication Date:
08 May 2020 (online)

Abstract

Background Emicizumab prophylaxis is a promising treatment that reduces bleeding events in severely affected patients with hemophilia A (PwHA). It is anticipated that emicizumab could be similarly effective in mild/moderate PwHA (PwMHA) although this effect has not been investigated.

Aim We evaluated ex vivo coagulant effects of emicizumabin PwMHA.

Methods Clot waveform analysis (CWA) triggered by prothrombin time/activated partial prothrombin time-mixed reagents was utilized to examine coagulant effects of emicizumabin factor (F)VIII-deficient plasma mixed with recombinant (r)FVIIIand in native plasmas from 16 PwMHA. The CWA parameter, adjusted-|min1| (Ad|min1|), was used. Increases in Ad|min1| (ΔAd|min1|) mediated by emicizumab were calculated from the slopes of regression lines in the presence of rFVIII.

Results Ad|min1| in FVIII-deficient plasma with various concentrations of rFVIII negatively correlated with ΔAd|min1|by adding emicizumab, and these data were defined as standard reference values. Ad|min1| (4.57 ± 0.50) in 16 PwMHA increased to 5.05 ± 0.54 and 5.37 ± 0.60 by adding emicizumab at 50 and 100 μg/mL, respectively, but remained lower than the normal range (7.22 ± 0.21). ΔAd|min1| levels were 1.5 to 2-fold higher in five cases and 0.4 to 0.6-fold lower in four cases, compared with reference values determined by rFVIII. In some cases, genetic analyses suggested that specific point mutations could have contributed to these findings. Further studies using rFVIII mutants indicated, however, that the differences in ΔAd|min1| were not related to individual FVIII gene defects.

Conclusion Emicizumab enhances coagulation potential in PwMHA. Assessment of ex vivo coagulant activity of emicizumab could be helpful for predicting coagulant potentials prior to treatment in these patients.

Authors' Contributions

Y.N. performed experiments and prepared the FVIII mutants, analyzed the data, prepared figures, and wrote the manuscript. K.N. designed all experiments, interpreted the data, prepared figures, wrote and edited the manuscript, and approved the final version to be published. K.Y. performed gene analyses and interpreted the data. S.F. performed gene analyses and prepared the FVIII mutants. M.N.S. and M.H. supervised this study and prepared emicizumab. M.S. supervised this study.


Note

An account of this work was presented at the ISTH 2019 Congress, Melbourne, Australia.


Supplementary Material

 
  • References

  • 1 Luck Jr JV, Silva M, Rodriguez-Merchan EC, Ghalambor N, Zahiri CA, Finn RS. Hemophilic arthropathy. J Am Acad Orthop Surg 2004; 12 (04) 234-245
  • 2 White II GC, Rosendaal F, Aledort LM, Lusher JM, Rothschild C, Ingerslev J. ; Factor VIII and Factor IX Subcommittee. Definitions in hemophilia. Recommendation of the scientific subcommittee on factor VIII and factor IX of the scientific and standardization committee of the International Society on Thrombosis and Haemostasis. Thromb Haemost 2001; 85 (03) 560
  • 3 Srivastava A, Brewer AK, Mauser-Bunschoten EP. , et al; Treatment Guidelines Working Group on Behalf of The World Federation Of Hemophilia. Guidelines for the management of hemophilia. Haemophilia 2013; 19 (01) e1-e47
  • 4 Ling M, Heysen JP, Duncan EM, Rodgers SE, Lloyd JV. High incidence of ankle arthropathy in mild and moderate haemophilia A. Thromb Haemost 2011; 105 (02) 261-268
  • 5 Di Minno MN, Ambrosino P, Franchini M, Coppola A, Di Minno G. Arthropathy in patients with moderate hemophilia a: a systematic review of the literature. Semin Thromb Hemost 2013; 39 (07) 723-731
  • 6 Beeton K, Neal D, Watson T, Lee CA. Parents of children with haemophilia--a transforming experience. Haemophilia 2007; 13 (05) 570-579
  • 7 Witmer C, Young G. Factor VIII inhibitors in hemophilia A: rationale and latest evidence. Ther Adv Hematol 2013; 4 (01) 59-72
  • 8 Caram C, de Souza RG, de Sousa JC. , et al. The long-term course of factor VIII inhibitors in patients with congenital haemophilia A without immune tolerance induction. Thromb Haemost 2011; 105 (01) 59-65
  • 9 Margaglione M, Castaman G, Morfini M. , et al; AICE-Genetics Study Group. The Italian AICE-Genetics hemophilia A database: results and correlation with clinical phenotype. Haematologica 2008; 93 (05) 722-728
  • 10 Eckhardt CL, van Velzen AS, Peters M. , et al; INSIGHT Study Group. Factor VIII gene (F8) mutation and risk of inhibitor development in nonsevere hemophilia A. Blood 2013; 122 (11) 1954-1962
  • 11 Kitazawa T, Igawa T, Sampei Z. , et al. A bispecific antibody to factors IXa and X restores factor VIII hemostatic activity in a hemophilia A model. Nat Med 2012; 18 (10) 1570-1574
  • 12 Sampei Z, Igawa T, Soeda T. , et al. Identification and multidimensional optimization of an asymmetric bispecific IgG antibody mimicking the function of factor VIII cofactor activity. PLoS ONE 2013; 8 (02) e57479
  • 13 Nogami K, Soeda T, Matsumoto T, Kawabe Y, Kitazawa T, Shima M. Routine measurements of factor VIII activity and inhibitor titer in the presence of emicizumab utilizing anti-idiotype monoclonal antibodies. J Thromb Haemost 2018; 16 (07) 1383-1390
  • 14 Nogami K, Matsumoto T, Tabuchi Y. , et al. Modified clot waveform analysis to measure plasma coagulation potential in the presence of the anti-factor IXa/factor X bispecific antibody emicizumab. J Thromb Haemost 2018; 16 (06) 1078-1088
  • 15 Shima M, Hanabusa H, Taki M. , et al. Factor VIII-mimetic function of humanized bispecific antibody in hemophilia A. N Engl J Med 2016; 374 (21) 2044-2053
  • 16 Shima M, Hanabusa H, Taki M. , et al. Long-term safety and efficacy of emicizumab in a phase 1/2 study in patients with hemophilia A with or without inhibitors. Blood Adv 2017; 1 (22) 1891-1899
  • 17 Oldenburg J, Mahlangu JN, Kim B. , et al. Emicizumab Prophylaxis in Hemophilia A with Inhibitors. N Engl J Med 2017; 377 (09) 809-818
  • 18 Young G, Liesner R, Sidonio RF. , et al. Emicizumab prophylaxis provides flexible and effective bleed control in children with hemophilia A with inhibitors: results from the HAVEN 2 study. Blood 2018; 132 (Suppl. 01) 632
  • 19 Mahlangu J, Oldenburg J, Paz-Priel I. , et al. Emicizumab prophylaxis in patients who have hemophilia A without inhibitors. N Engl J Med 2018; 379 (09) 811-822
  • 20 Pipe SW, Shima M, Lehle M. , et al. Efficacy, safety, and pharmacokinetics of emicizumab prophylaxis given every 4 weeks in people with haemophilia A (HAVEN 4): a multicentre, open-label, non-randomised phase 3 study. Lancet Haematol 2019; 6 (06) e295-e305
  • 21 Shima M, Scandella D, Yoshioka A. , et al. A factor VIII neutralizing monoclonal antibody and a human inhibitor alloantibody recognizing epitopes in the C2 domain inhibit factor VIII binding to von Willebrand factor and to phosphatidylserine. Thromb Haemost 1993; 69 (03) 240-246
  • 22 Williams IJ, Abuzenadah A, Winship PR. , et al. Precise carrier diagnosis in families with haemophilia A: use of conformation sensitive gel electrophoresis for mutation screening and polymorphism analysis. Thromb Haemost 1998; 79 (04) 723-726
  • 23 Nogami K, Zhou Q, Wakabayashi H, Fay PJ. Thrombin-catalyzed activation of factor VIII with His substituted for Arg372 at the P1 site. Blood 2005; 105 (11) 4362-4368
  • 24 Potgieter JJ, Damgaard M, Hillarp A. One-stage vs. chromogenic assays in haemophilia A. Eur J Haematol 2015; 94 (Suppl. 77) 38-44
  • 25 Eaton D, Rodriguez H, Vehar GA. Proteolytic processing of human factor VIII. Correlation of specific cleavages by thrombin, factor Xa, and activated protein C with activation and inactivation of factor VIII coagulant activity. Biochemistry 1986; 25 (02) 505-512
  • 26 Newell JL, Fay PJ. Cleavage at Arg-1689 influences heavy chain cleavages during thrombin-catalyzed activation of factor VIII. J Biol Chem 2009; 284 (17) 11080-11089
  • 27 den Uijl IE, Fischer K, Van Der Bom JG, Grobbee DE, Rosendaal FR, Plug I. Analysis of low frequency bleeding data: the association of joint bleeds according to baseline FVIII activity levels. Haemophilia 2011; 17 (01) 41-44
  • 28 Muto A, Yoshihashi K, Takeda M. , et al. Anti-factor IXa/X bispecific antibody (ACE910): hemostatic potency against ongoing bleeds in a hemophilia A model and the possibility of routine supplementation. J Thromb Haemost 2014; 12 (02) 206-213
  • 29 Shima M, Matsumoto T, Fukuda K. , et al. The utility of activated partial thromboplastin time (aPTT) clot waveform analysis in the investigation of hemophilia A patients with very low levels of factor VIII activity (FVIII:C). Thromb Haemost 2002; 87 (03) 436-441
  • 30 Matsumoto T, Nogami K, Tabuchi Y. , et al. Clot waveform analysis using CS-2000i™ distinguishes between very low and absent levels of factor VIII activity in patients with severe haemophilia A. Haemophilia 2017; 23 (05) e427-e435
  • 31 Peters RT, Toby G, Lu Q. , et al. Biochemical and functional characterization of a recombinant monomeric factor VIII-Fc fusion protein. J Thromb Haemost 2013; 11 (01) 132-141
  • 32 Nogami K, Shima M. Phenotypic heterogeneity of hemostasis in severe hemophilia. Semin Thromb Hemost 2015; 41 (08) 826-831
  • 33 Pipe SW, Eickhorst AN, McKinley SH, Saenko EL, Kaufman RJ. Mild hemophilia A caused by increased rate of factor VIII A2 subunit dissociation: evidence for nonproteolytic inactivation of factor VIIIa in vivo. Blood 1999; 93 (01) 176-183
  • 34 Yada K, Nogami K, Wakabayashi H, Fay PJ, Shima M. The mild phenotype in severe hemophilia A with Arg1781His mutation is associated with enhanced binding affinity of factor VIII for factor X. Thromb Haemost 2013; 109 (06) 1007-1015
  • 35 EAHAD. F8 variant database. Available at: http://f8-db.eahad.org/ . Accessed April 13, 2020
  • 36 Kitazawa T, Esaki K, Tachibana T. , et al. Factor VIIIa-mimetic cofactor activity of a bispecific antibody to factors IX/IXa and X/Xa, emicizumab, depends on its ability to bridge the antigens. Thromb Haemost 2017; 117 (07) 1348-1357