Thromb Haemost
DOI: 10.1055/a-2315-8199
Coagulation and Fibrinolysis

Additional Factor X Enhances Emicizumab-Driven Coagulation Function in Patients with Hemophilia A and Hemophilia A Mice

Kazuki Shimizu
1   Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
,
Yuto Nakajima
1   Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
2   Advanced Medical Science of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Nara, Japan
,
Eisuke Takami
3   Medical Affairs Section, KM Biologics Co., Ltd, Kumamoto, Japan
,
Hirotoshi Nakano
3   Medical Affairs Section, KM Biologics Co., Ltd, Kumamoto, Japan
,
Keiji Nogami
1   Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
› Institutsangaben
Funding This research was supported, in part, by KM Biologics Co., Ltd. And Grant-in-Aid for Scientific Research (KAKENHI) from the Ministry of Education, Culture, Sports, Science and Technology (MEXT) to K.N. (21K07804) and Y.N. (22K15928).


Abstract

Background Bypassing agents are used for breakthrough bleedings in patients with hemophilia A with inhibitor (PwHAwI) receiving emicizumab prophylaxis. Previous study demonstrated a weak binding affinity between emicizumab and factor (F)X (K d; 1.85 μM), and that this value was much greater than the plasma FX concentration (∼130 nM). We speculated that increased FX levels could enhance coagulation potential in emicizumab-treated patients with hemophilia A (PwHA). To investigate the relationship between FX concentrations and emicizumab-driven coagulation.

Methods Plasma FX (up to 1,040 nM) and emicizumab (50 µg/mL) were added to FVIII-deficient plasmas, and plasma-derived FX (520 nM) or recombinant (r)FVIIa (2.2 µg/mL) was added to plasmas from three emicizumab-treated PwHAwI. The adjusted maximum coagulation velocity (Ad|min1|) by clot waveform analysis and peak thrombin (PeakTh) by thrombin generation assay in them were evaluated. Emicizumab (3.0 mg/kg), human (h)FIX (100 IU/kg), and various doses of hFX (100–500 IU/kg) were intravenously administered to HA mice. Clotting time/clot formation time (CT/CFT) were assessed using rotational thromboelastometry, and blood loss was estimated by a tail-clip assay.

Results The addition of FX to FVIII-deficient plasma with emicizumab increased Ad|min1| and PeakTh. The coagulation parameters in emicizumab-treated PwHAwI spiked with additional FX remained within the normal range as well as the additional rFVIIa. In animal models, hFX injection shortened the CT and CT + CFT. The shorter CT and CT + CFT, and the lower blood loss were evident after 200 or 500 IU/kg hFX administration, and those indices were comparable to those in wild-type mice.

Conclusion Supplementation with FX may improve emicizumab-driven hemostasis in PwHA.

Note

An account of this work was presented at the annual meeting of ISTH2023 Congress, Montréal, Quebec, Canada.


Data Availability Statement

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.


Authors' Contribution

K.S. performed the experiments, analyzed the data, made the figures, and wrote the paper. Y.N. designed all experiments, interpreted the data, prepared figures, wrote and edited the manuscript, and approved the final version to be published. E.T. and H.N. supervised the study and prepared pd-FVIIa/FX, FIX, and FX concentrates. K.N. designed the research, supported the data, interpreted the data, wrote the paper, and edited the manuscript.


Supplementary Material



Publikationsverlauf

Eingereicht: 06. Dezember 2023

Angenommen: 25. April 2024

Accepted Manuscript online:
27. April 2024

Artikel online veröffentlicht:
15. Mai 2024

© 2024. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Gouw SC, van der Bom JG, Ljung R. et al; PedNet and RODIN Study Group. Factor VIII products and inhibitor development in severe hemophilia A. N Engl J Med 2013; 368 (03) 231-239
  • 2 Walsh CE, Jiménez-Yuste V, Auerswald G, Grancha S. The burden of inhibitors in haemophilia patients. Thromb Haemost 2016; 116 (Suppl. 01) S10-S17
  • 3 Astermark J, Rocino A, Von Depka M. et al; EHTSB. Current use of by-passing agents in Europe in the management of acute bleeds in patients with haemophilia and inhibitors. Haemophilia 2007; 13 (01) 38-45
  • 4 Berntorp E. Differential response to bypassing agents complicates treatment in patients with haemophilia and inhibitors. Haemophilia 2009; 15 (01) 3-10
  • 5 Ogiwara K, Nogami K, Matsumoto T, Shima M. Tissue factor pathway inhibitor in activated prothrombin complex concentrates (aPCC) moderates the effectiveness of therapy in some severe hemophilia A patients with inhibitor. Int J Hematol 2014; 99 (05) 577-587
  • 6 Tomokiyo K, Nakatomi Y, Araki T. et al. A novel therapeutic approach combining human plasma-derived Factors VIIa and X for haemophiliacs with inhibitors: evidence of a higher thrombin generation rate in vitro and more sustained haemostatic activity in vivo than obtained with Factor VIIa alone. Vox Sang 2003; 85 (04) 290-299
  • 7 Nakatomi Y, Nakashima T, Gokudan S. et al. Combining FVIIa and FX into a mixture which imparts a unique thrombin generation potential to hemophilic plasma: an in vitro assessment of FVIIa/FX mixture as an alternative bypassing agent. Thromb Res 2010; 125 (05) 457-463
  • 8 Shirahata A, Fukutake K, Takamatsu J. et al. A phase II clinical trial of a mixture of plasma-derived factor VIIa and factor X (MC710) in haemophilia patients with inhibitors: haemostatic efficacy, safety and pharmacokinetics/pharmacodynamics. Haemophilia 2013; 19 (06) 853-860
  • 9 Shinkoda Y, Shirahata A, Fukutake K. et al. A phase III clinical trial of a mixture agent of plasma-derived factor VIIa and factor X (MC710) in haemophilia patients with inhibitors. Haemophilia 2017; 23 (01) 59-66
  • 10 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
  • 11 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
  • 12 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
  • 13 Shima M, Hanabusa H, Taki M. et al. Long-term safety and efficacy of emicizumab in a phase 1/2 study in hemophilia A patients with or without inhibitors. Blood Adv 2017; 1 (22) 1891-1899
  • 14 Oldenburg J, Mahlangu JN, Kim B. et al. Emicizumab prophylaxis in hemophilia A with inhibitors. N Engl J Med 2017; 377 (09) 809-818
  • 15 Young G, Liesner R, Chang T. et al. A multicenter, open-label phase 3 study of emicizumab prophylaxis in children with hemophilia A with inhibitors. Blood 2019; 134 (24) 2127-2138
  • 16 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
  • 17 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
  • 18 Shima M, Nogami K, Nagami S. et al. A multicentre, open-label study of emicizumab given every 2 or 4 weeks in children with severe haemophilia A without inhibitors. Haemophilia 2019; 25 (06) 979-987
  • 19 Nakajima Y, Mizumachi K, Shimonishi N. et al. Comparisons of global coagulation potential and bleeding episodes in emicizumab-treated hemophilia A patients and mild hemophilia A patients. Int J Hematol 2022; 115 (04) 489-498
  • 20 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
  • 21 Kizilocak H, Marquez-Casas E, Phei Wee C, Malvar J, Carmona R, Young G. Comparison of bypassing agents in patients on emicizumab using global hemostasis assays. Haemophilia 2021; 27 (01) 164-172
  • 22 Hertzberg M. Biochemistry of factor X. Blood Rev 1994; 8 (01) 56-62
  • 23 Mann KG, Nesheim ME, Church WR, Haley P, Krishnaswamy S. Surface-dependent reactions of the vitamin K-dependent enzyme complexes. Blood 1990; 76 (01) 1-16
  • 24 Kamikubo Y, Mendolicchio GL, Zampolli A. et al. Selective factor VIII activation by the tissue factor-factor VIIa-factor Xa complex. Blood 2017; 130 (14) 1661-1670
  • 25 Furukawa S, Nogami K, Ogiwara K, Shima M. Potential role of activated factor VIII (FVIIIa) in FVIIa/tissue factor-dependent FXa generation in initiation phase of blood coagulation. Int J Hematol 2019; 109 (04) 390-401
  • 26 Louvain-Quintard VB, Bianchini EP, Calmel-Tareau C, Tagzirt M, Le Bonniec BF. Thrombin-activable factor X re-establishes an intrinsic amplification in tenase-deficient plasmas. J Biol Chem 2005; 280 (50) 41352-41359
  • 27 Bunce MW, Toso R, Camire RM. Zymogen-like factor Xa variants restore thrombin generation and effectively bypass the intrinsic pathway in vitro. Blood 2011; 117 (01) 290-298
  • 28 Muczynski V, Verhenne S, Casari C. et al. A thrombin-activatable factor X variant corrects hemostasis in a mouse model for hemophilia A. Thromb Haemost 2019; 119 (12) 1981-1993
  • 29 Abache T, Fontayne A, Grenier D. et al. A mutated factor X activatable by thrombin corrects bleedings in vivo in a rabbit model of antibody-induced hemophilia A. Haematologica 2020; 105 (09) 2335-2340
  • 30 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
  • 31 Cooper DN, Millar DS, Wacey A, Pemberton S, Tuddenham EG. Inherited factor X deficiency: molecular genetics and pathophysiology. Thromb Haemost 1997; 78 (01) 161-172
  • 32 Okuda M, Yamamoto Y. Usefulness of synthetic phospholipid in measurement of activated partial thromboplastin time: a new preparation procedure to reduce batch difference. Clin Lab Haematol 2004; 26 (03) 215-223
  • 33 Nakajima Y, Takami E, Nakano H, Nogami K. In vitro evaluation of global coagulation potentials in the co-presence of plasma-derived factors Viia/X products (Byclot® ) and emicizumab in patients with haemophilia A and inhibitors and acquired haemophilia A: a pilot study. Haemophilia 2022; 28 (05) e149-e152
  • 34 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
  • 35 Nakajima Y, Takeyama M, Oda A, Shimonishi N, Nogami K. Factor VIII mutated with Lys1813Ala within the factor IXa-binding region enhances intrinsic coagulation potential. Blood Adv 2023; 7 (08) 1436-1445
  • 36 Ferrière S, Peyron I, Christophe OD. et al. A hemophilia A mouse model for the in vivo assessment of emicizumab function. Blood 2020; 136 (06) 740-748
  • 37 Collins PW, Liesner R, Makris M. et al. Treatment of bleeding episodes in haemophilia A complicated by a factor VIII inhibitor in patients receiving Emicizumab. Interim guidance from UKHCDO Inhibitor Working Party and Executive Committee. Haemophilia 2018; 24 (03) 344-347
  • 38 Yada K, Nogami K, Ogiwara K. et al. Global coagulation function assessed by rotational thromboelastometry predicts coagulation-steady state in individual hemophilia A patients receiving emicizumab prophylaxis. Int J Hematol 2019; 110 (04) 419-430
  • 39 Ogiwara K, Nogami K, Matsumoto N. et al. A modified thrombin generation assay to evaluate the plasma coagulation potential in the presence of emicizumab, the bispecific antibody to factors IXa/X. Int J Hematol 2020; 112 (05) 621-630
  • 40 Gilbert GE, Arena AA. Activation of the factor VIIIa-factor IXa enzyme complex of blood coagulation by membranes containing phosphatidyl-L-serine. J Biol Chem 1996; 271 (19) 11120-11125
  • 41 Mak S, Marszal A, Matscheko N, Rant U. Kinetic analysis of ternary and binary binding modes of the bispecific antibody emicizumab. MAbs 2023; 15 (01) 2149053
  • 42 Frostell A, Vinterbäck L, Sjöbom H. Protein-ligand interactions using SPR systems. Methods Mol Biol 2013; 1008: 139-165
  • 43 Bolliger D, Szlam F, Molinaro RJ, Rahe-Meyer N, Levy JH, Tanaka KA. Finding the optimal concentration range for fibrinogen replacement after severe haemodilution: an in vitro model. Br J Anaesth 2009; 102 (06) 793-799
  • 44 Kizilocak H, Yukhtman CL, Marquez-Casas E, Lee J, Donkin J, Young G. Management of perioperative hemostasis in a severe hemophilia A patient with inhibitors on emicizumab using global hemostasis assays. Ther Adv Hematol 2019; 10: 2040620719860025
  • 45 Villar A, Aronis S, Morfini M. et al. Pharmacokinetics of activated recombinant coagulation factor VII (NovoSeven) in children vs. adults with haemophilia A. Haemophilia 2004; 10 (04) 352-359
  • 46 Roberts HR, Lechler E, Webster WP, Penick GD. Survival of transfused factor X in patients with stuart disease. Thromb Diath Haemorrh 1965; 13: 305-313
  • 47 Simioni P, Tormene D, Tognin G. et al. X-linked thrombophilia with a mutant factor IX (factor IX Padua). N Engl J Med 2009; 361 (17) 1671-1675