1
|
Shimizu K, Nakajima Y, Takami E, Nakano H, Nogami K. Additional Factor X Enhances Emicizumab-Driven Coagulation Function in Patients with Hemophilia A and Hemophilia A Mice. Thromb Haemost 2025; 125:21-32. [PMID: 38677278 DOI: 10.1055/a-2315-8199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
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.
Collapse
Affiliation(s)
- Kazuki Shimizu
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | - Yuto Nakajima
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
- Advanced Medical Science of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Nara, Japan
| | - Eisuke Takami
- Medical Affairs Section, KM Biologics Co., Ltd, Kumamoto, Japan
| | | | - Keiji Nogami
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
2
|
Nakajima Y, Osuna M, Mizumachi K, Shimonishi N, Furukawa S, Ogiwara K, Nogami K. Activated protein C resistance in the copresence of emicizumab and activated prothrombin complex concentrates. Res Pract Thromb Haemost 2024; 8:102479. [PMID: 39114481 PMCID: PMC11305313 DOI: 10.1016/j.rpth.2024.102479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/11/2024] [Accepted: 06/12/2024] [Indexed: 08/10/2024] Open
Abstract
Background Venous thromboembolic events have been reported in persons with hemophilia A who received emicizumab and activated prothrombin complex concentrate (APCC) concomitantly, but the relevant mechanism(s) remains unclear. We speculated that activated protein C (APC) and antithrombin (AT) resistance might be associated with these adverse events. Objectives To investigate APC and AT resistance in factor (F)VIII-deficient (FVIIIdef) plasma in the presence of emicizumab and APCC. Methods In pooled normal plasma or FVIIIdef plasma samples mixed with emicizumab (50 μg/mL) and FVIII-bypassing agents, including recombinant FVIIa (2.2 μg/mL), APCC (1.3 IU/mL), or plasma-derived FVIIa/FX (1.5 μg/mL), the suppression effect of AT (0-2.4 μM) and APC (0-16 nM) was assessed by tissue factor-triggered thrombin generation assay. The APC effects in FVIIIdef plasma with the copresence of emicizumab, FII (1.3 μM), and/or FIXa (280 pM) were also examined. Results The AT resistance in emicizumab and each bypassing agent was not observed. Moreover, APC dose-dependent suppression effect was observed in pooled normal plasma or FVIIIdef plasma mixed with emicizumab and recombinant FVIIa or plasma-derived FVIIa/FX. However, APC-catalyzed inactivation had little effect on thrombin generation assay potential in FVIIIdef plasma spiked with emicizumab and APCC. The addition of FIXa to emicizumab in FVIIIdef plasma could lead to partial APC resistance. Furthermore, FVIIIdef plasma spiked with emicizumab, FIXa, and FII was markedly resistant to APC-mediated inactivation. Conclusion FII and FIXa in APCCs were key clotting factors for APC resistance in FVIIIdef plasma supplemented with emicizumab and APCCs. The APC resistance in persons with hemophilia A receiving emicizumab and APCC may contribute to venous thromboembolic events.
Collapse
Affiliation(s)
- Yuto Nakajima
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
- Advanced Medical Science of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Nara, Japan
| | - Mitsumasa Osuna
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | | | - Naruto Shimonishi
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
- The Course of Thrombosis and Hemostasis Molecular Pathology, Nara Medical University, Kashihara, Nara, Japan
| | - Shoko Furukawa
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | - Kenichi Ogiwara
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | - Keiji Nogami
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
3
|
Sakai M, Amano K, Chin M, Takedani H, Ishida H, Sakashita K, Taki M, Migita M, Watanabe H, Ishimura M, Nogami K, Harano S, Shirahata A. Plasma-derived factor VIIa and factor X mixture agent (MC710) prophylaxis in haemophilia B patients with inhibitors. Haemophilia 2023; 29:456-465. [PMID: 36516312 DOI: 10.1111/hae.14710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Haemophilia B patients with factor IX inhibitors have particularly unmet needs for conventional therapy. AIM Phase II/III clinical trial, multicentre, open-label, prospective, self-controlled study was conducted to assess MC710 prophylaxis in haemophilia B patients with inhibitors. METHODS We enrolled haemophilia patients who had received episodic or prophylactic treatment with bypassing agents up to that time. The participants continued their conventional therapy for 24 weeks and then MC710 was prophylactically infused intravenously every 2 or 3 days at 60 to 120 μg as FVIIa per kilogram of body weight for 24 weeks. The primary endpoint was the annual bleeding rate (ABR) requiring bypassing agents, which was compared intraindividually between the conventional therapy period and the MC710 prophylaxis period. RESULTS A total of 11 male haemophilia B patients were enrolled. The median ABR ratio for each participant (the prophylaxis period ABR divided by the conventional therapy period ABR) was .33 (2.1/6.5), range from .00 to 3.77. ABR ratios for 9 of the 11 patients ranged from .00 to .60, and 3 of the 9 patients had zero bleeding events during the prophylaxis period. Meanwhile, ABR ratios for the remaining two patients were 2.53 and 3.77, respectively. Although a fibrinogen decrease recovered by the dose reduction was reported for only one participant as the sole adverse drug reaction in this study, no thrombotic events or other safety concerns were reported. CONCLUSION MC710 prophylaxis is considered to be decrease the bleeding rate in haemophilia B patients with inhibitors without safety concerns.
Collapse
Affiliation(s)
- Michio Sakai
- Department of Pediatrics, Munakata Suikokai General Hospital, Fukuoka, Japan
| | - Kagehiro Amano
- Department of Laboratory Medicine, Tokyo Medical University Hospital, Tokyo, Japan
| | - Motoaki Chin
- Department of Pediatrics, Nihon University Itabashi Hospital, Tokyo, Japan
| | - Hideyuki Takedani
- Department of Joint Surgery, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | - Kazuo Sakashita
- Department of Hematology Oncology, Nagano Children's Hospital, Nagano, Japan
| | - Masashi Taki
- Department of Pediatrics, St. Marianna University Hospital, Kanagawa, Japan
| | - Masahiro Migita
- Department of Pediatrics, Japanese Red Cross Kumamoto Hospital, Kumamoto, Japan
| | - Hiroyoshi Watanabe
- Department of Pediatrics, Tokushima University Hospital, Tokushima, Japan
| | - Masataka Ishimura
- Department of Pediatrics, Kyushu University Hospital, Fukuoka, Japan
| | - Keiji Nogami
- Department of Pediatrics, Nara Medical University, Nara, Japan
| | - Sho Harano
- Department of Clinical Development, KM Biologics Co., Ltd., Kumamoto, Japan
| | | |
Collapse
|
4
|
Takami E, Hashimoto K, Kitano R, Nishikawa K, Fuchigami T, Nakano H. Additive effect of factor X on the structure and stability of activated factor VII-induced fibrin clot in hemophilic plasma with inhibitor. Thromb Res 2023; 223:127-130. [PMID: 36739807 DOI: 10.1016/j.thromres.2023.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/11/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023]
Affiliation(s)
- Eisuke Takami
- Medical Affairs Section, KM Biologics Co., Ltd, Kumamoto, Japan.
| | - Kohei Hashimoto
- Technical Development Department, KM Biologics Co., Ltd, Kumamoto, Japan
| | - Ryoichi Kitano
- Technical Development Department, KM Biologics Co., Ltd, Kumamoto, Japan
| | - Kazumi Nishikawa
- Technical Development Department, KM Biologics Co., Ltd, Kumamoto, Japan
| | - Takashi Fuchigami
- Technical Development Department, KM Biologics Co., Ltd, Kumamoto, Japan
| | | |
Collapse
|
5
|
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:e149-e152. [PMID: 35947585 DOI: 10.1111/hae.14650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/16/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Yuto Nakajima
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan.,Advanced Medical Science of Thrombosis and Hemostasis, Nara Medical University, Kashihara, Nara, Japan
| | - Eisuke Takami
- Medical Affairs Section, KM Biologics Co., Ltd, Kumamoto, Japan
| | | | - Keiji Nogami
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
6
|
Effects of low and high factor X concentrations on thrombin generation in vitro. THROMBOSIS UPDATE 2022. [DOI: 10.1016/j.tru.2022.100111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
7
|
Abstract
Haemophilia A and B are rare congenital, recessive X-linked disorders caused by lack or deficiency of clotting factor VIII (FVIII) or IX (FIX), respectively. The severity of the disease depends on the reduction of levels of FVIII or FIX, which are determined by the type of the causative mutation in the genes encoding the factors (F8 and F9, respectively). The hallmark clinical characteristic, especially in untreated severe forms, is bleeding (spontaneous or after trauma) into major joints such as ankles, knees and elbows, which can result in the development of arthropathy. Intracranial bleeds and bleeds into internal organs may be life-threatening. The median life expectancy was ~30 years until the 1960s, but improved understanding of the disorder and development of efficacious therapy based on prophylactic replacement of the missing factor has caused a paradigm shift, and today individuals with haemophilia can look forward to a virtually normal life expectancy and quality of life. Nevertheless, the potential development of inhibitory antibodies to infused factor is still a major hurdle to overcome in a substantial proportion of patients. Finally, gene therapy for both types of haemophilia has progressed remarkably and could soon become a reality.
Collapse
|
8
|
Uchida E, Komori K, Kurata T, Taki M, Sakashita K. Prophylaxis Using a Mixture of Plasma-Derived Activated Factor VII and Factor X (pdFVIIa/FX) in a Patient with Hemophilia B Complicated by Inhibitors and Allergy to Factor IX Concentrates: A Case Report. Acta Haematol 2020; 144:293-296. [PMID: 32702700 DOI: 10.1159/000508722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 05/17/2020] [Indexed: 12/31/2022]
Abstract
Treating patients with hemophilia and inhibitors is often problematic. The presence of inhibitors negatively impacts the effectiveness of treatment to achieve hemostasis especially in patients with hemophilia B, owing mainly to allergic reactions to factor IX (FIX) concentrates and the low success rate of immune tolerance therapy. A 9-month-old boy had intracranial hemorrhage and was diagnosed with hemophilia B. After replacement therapy, he developed inhibitors and an allergic reaction to FIX. Prophylactic therapy was initiated with recombinant activated factor VII (rFVIIa) and later switched to pdFVIIa/factor X (FX; 120 μg/kg as the FVII dose, every other day) because of a recurrence of intracranial hemorrhage. Since then, he remained well without life-threatening bleeding for more than 2 years. Our case suggests that pdFVIIa/FX may be useful for prophylactic therapy in hemophilia B complicated by inhibitors and allergic reaction to FIX concentrates.
Collapse
Affiliation(s)
- Eriko Uchida
- Department of Hematology and Oncology, Nagano Children's Hospital, Azumino, Japan
| | - Kazutoshi Komori
- Department of Hematology and Oncology, Nagano Children's Hospital, Azumino, Japan
| | - Takashi Kurata
- Department of Hematology and Oncology, Nagano Children's Hospital, Azumino, Japan
| | - Masashi Taki
- Department of Pediatrics, St. Marianna University School of Medicine Yokohama City Seibu Hospital, Yokohama, Japan
| | - Kazuo Sakashita
- Department of Hematology and Oncology, Nagano Children's Hospital, Azumino, Japan,
| |
Collapse
|
9
|
Ochi S, Takeyama M, Shima M, Nogami K. Plasma-derived factors VIIa and X mixtures (Byclot®) significantly improve impairment of coagulant potential ex vivo in plasmas from acquired hemophilia A patients. Int J Hematol 2020; 111:779-785. [DOI: 10.1007/s12185-020-02837-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 10/25/2022]
|
10
|
Takeshita S, Ogawa S, Nakayama Y, Mukai N, Nakajima Y, Mizobe T, Sawa T, Tanaka KA. Prohemostatic Activity of Factor X in Combination With Activated Factor VII in Dilutional Coagulopathy. Anesth Analg 2018; 129:339-345. [PMID: 30320649 DOI: 10.1213/ane.0000000000003858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Recombinant activated factor VII (rFVIIa) concentrate reduces allogeneic blood transfusions, but it may increase thromboembolic complications in complex cardiac surgery. The mixture of activated factor VII (FVIIa) and factor X (FX) (FVIIa/FX) (FVIIa:FX = 1:10) is a novel bypassing agent for hemophilia patients. We hypothesized that the combination of FX and FVIIa could improve thrombin generation (TG) in acquired multifactorial coagulation defects such as seen in cardiac surgery and conducted in vitro evaluation of FVIIa/FX in parallel with other coagulation factor concentrates using in vitro and in vivo diluted plasma samples. METHODS Plasma samples were collected from 9 healthy volunteers and 12 cardiac surgical patients. We measured TG (Thrombinoscope) using in vitro 50% dilution plasma and in vivo dilution plasma after cardiopulmonary bypass, in parallel with thromboelastometry (ROTEM) and standard coagulation assays. In vitro additions of FVIIa/FX (0.35, 0.7, and 1.4 μg/mL, based on the FVIIa level), rFVIIa (1.4, 2.8, and 6.4 μg/mL), prothrombin complex concentrate (0.3 international unit), and 20% plasma replacement were evaluated. RESULTS In diluted plasma, the addition of either FVIIa/FX or rFVIIa shortened the lag time and increased the peak TG, but the effect in lag time of FVIIa/FX at 0.35 μg/mL was more extensive than rFVIIa at 6.4 μg/mL. Prothrombin complex concentrate increased peak TG by increasing the prothrombin level but failed to shorten the lag time. No improvement in any of the TG variables was observed after 20% volume replacement with plasma. The addition of factor concentrates normalized prothrombin time/international normalized ratio but not with plasma replacement. In cardiac patients, similar patterns were observed on TG in post-cardiopulmonary bypass samples. FVIIa/FX shortened clotting time (CT) in a concentration-dependent manner on CT on thromboelastometry. Plasma replacement did not improve CT, but a combination of plasma and FVIIa/FX (0.35 μg/mL) more effectively shortened CT than FVIIa/FX alone. CONCLUSIONS The combination of FVIIa and FX improved TG more efficiently than rFVIIa alone or plasma in dilutional coagulopathy models. The required FVIIa dose in FVIIa/FX was considerably lower than those reported during bypassing therapy in hemophilia patients (1.4-2.8 μg/mL). The combination of plasma could restore coagulation more efficiently compared to FVIIa/FX alone. Lesser FVIIa requirement to exert procoagulant activity may be favorable in terms of reducing systemic thromboembolic complications.
Collapse
Affiliation(s)
- Shusuke Takeshita
- From the Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoru Ogawa
- From the Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshinobu Nakayama
- From the Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nobuhiro Mukai
- From the Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasufumi Nakajima
- Department of Anesthesiology, Kansai Medical University, Osaka, Japan
| | - Toshiki Mizobe
- From the Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Teiji Sawa
- From the Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenichi A Tanaka
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
11
|
Shinkoda Y, Shirahata A, Fukutake K, Takamatsu J, Shima M, Hanabusa H, Mugishima H, Takedani H, Kawasugi K, Taki M, Matsushita T, Tawa A, Nogami K, Higasa S, Kosaka Y, Fujii T, Sakai M, Migita M, Uchiba M, Kawakami K, Sameshima K, Ohashi Y, Saito H. A phase III clinical trial of a mixture agent of plasma-derived factor VIIa and factor X (MC710) in haemophilia patients with inhibitors. Haemophilia 2016; 23:59-66. [DOI: 10.1111/hae.13050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2016] [Indexed: 11/27/2022]
Affiliation(s)
- Y. Shinkoda
- Department of Paediatrics; Kagoshima City Hospital; Kagoshima Japan
| | - A. Shirahata
- Kitakyushu Yahata Higashi Hospital; Kitakyushu Fukuoka Japan
| | - K. Fukutake
- Department of Laboratory Medicine; Tokyo Medical University; Tokyo Japan
| | - J. Takamatsu
- Japanese Red Cross Tokai-Hokuriku Block Blood Center; Seto Aichi Japan
| | - M. Shima
- Department of Paediatrics; Nara Medical University; Kashihara Nara Japan
| | - H. Hanabusa
- Department of Haematology; Ogikubo Hospital; Tokyo Japan
| | - H. Mugishima
- Department of Paediatrics; Nihon University Itabashi Hospital; Tokyo Japan
| | - H. Takedani
- Department of Joint Surgery; Research Hospital of the Institute of Medical Science; the University of Tokyo; Tokyo Japan
| | - K. Kawasugi
- Department of Internal Medicine; Teikyo University Hospital; Tokyo Japan
| | - M. Taki
- Department of Paediatrics; St. Marianna University School of Medicine; Kawasaki Kanagawa Japan
| | - T. Matsushita
- Department of Transfusion Medicine; Nagoya University Hospital; Nagoya Aichi Japan
| | - A. Tawa
- Department of Paediatrics; National Hospital Organization Osaka National Hospital; Osaka Japan
| | - K. Nogami
- Department of Paediatrics; Nara Medical University; Kashihara Nara Japan
| | - S. Higasa
- Division of Haematology; Department of Internal Medicine; Hyogo College of Medicine; Nishinomiya Hyogo Japan
| | - Y. Kosaka
- Department of Haematology and Oncology; Kobe Children's Hospital; Kobe Hyogo Japan
| | - T. Fujii
- Division of Blood Transfusion; Hiroshima University Hospital; Hiroshima Japan
| | - M. Sakai
- Department of Paediatrics; University of Occupational and Environmental Health; Kitakyushu Fukuoka Japan
| | - M. Migita
- Department of Paediatrics; Kumamoto Red Cross Hospital; Kumamoto Japan
| | - M. Uchiba
- Department of Blood Transfusion and Cell Therapy; Kumamoto University Hospital; Kumamoto Japan
| | - K. Kawakami
- Department of Paediatrics; Kagoshima City Hospital; Kagoshima Japan
| | - K. Sameshima
- Department of Paediatrics; Kagoshima City Hospital; Kagoshima Japan
| | - Y. Ohashi
- Department of Biostatistics; School of Public Health; the University of Tokyo; Tokyo Japan
| | - H. Saito
- National Hospital Organization Nagoya Medical Center; Nagoya Aichi Japan
| |
Collapse
|
12
|
Shima M, Hermans C, de Moerloose P. Novel products for haemostasis. Haemophilia 2014; 20 Suppl 4:29-35. [PMID: 24762272 DOI: 10.1111/hae.12413] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2014] [Indexed: 01/16/2023]
Abstract
The primary major issue in haemophilia treatment remains the development of inhibitors. Recently two novel bypassing products have been developed. First, a humanized bispecific antibody against FIXa and FX, termed hBS23, was produced utilizing these two molecules placed into a spatially appropriate position to mimic FVIIIa, and recently this mimetic activity and the pharmacokinetics of the original antibody were improved by engineering the charge properties of the variable region within the immunoglobulin. Using the new antibody, termed ACE910, a phase 1 study in 64 Japanese and Caucasian healthy adults was performed and data from this trial suggested that the product had medically acceptable safety and tolerability profiles. The other new bypassing agent is named MC710, and consists of a mixture of plasma-derived FVIIa and FX. Preclinical studies using in vitro and in vivo haemophilia B inhibitor monkey models indicated that the haemostatic effects of FVIIa and FX were enhanced by simultaneous administration. Results from phase I and II clinical studies suggested that MC710 had equal or greater pharmacokinetic (PK), pharmacodynamic (PD), efficacy and safety profiles than conventional bypassing agents in the treatment of joint bleeding in haemophilia patients with inhibitors. Another significant current issue in this context is the increased medical cost of conventional treatment due to the higher consumption of concentrates. Biosimilar products may offer advantages in these circumstances and may offer a less expensive alternative. Regulatory issues, however, together with acceptability of biosimilar materials and reimbursement policies as well as supply and demand incentives remain to be considered. Rare bleeding disorders (RBDs) have attracted less attention from the pharmaceutical industry than haemophilia or von Willebrand disease due to the limited number of patients involved. Many cases of this type have been treated, therefore, using fresh frozen plasma (FFP) or prothrombin complex concentrates (PCCs) which carry serious risks of infections, allergic reactions and fluid overload. Several specific plasma-derived or recombinant products including fibrinogen, FVIIa, FXI and FXIII have now become available, however, and a phase III clinical study of recombinant FXIIIa has recently been completed demonstrating safety and efficacy of substances of this nature.
Collapse
Affiliation(s)
- M Shima
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan
| | | | | |
Collapse
|
13
|
Promising coagulation factor VIII bypassing strategies for patients with haemophilia A. Blood Coagul Fibrinolysis 2014; 25:539-52. [DOI: 10.1097/mbc.0000000000000098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
14
|
Shibeko AM, Woodle SA, Mahmood I, Jain N, Ovanesov MV. Predicting dosing advantages of factor VIIa variants with altered tissue factor-dependent and lipid-dependent activities. J Thromb Haemost 2014; 12:1302-12. [PMID: 24913469 DOI: 10.1111/jth.12628] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/28/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND Recombinant factor VIIa (rFVIIa) is an FX-cleaving coagulation enzyme licensed for the treatment of bleeding episodes in hemophiliacs with inhibitory antibodies. Even though the optimal dosing and comparative dose efficacy of rFVIIa remain poorly understood, genetic or chemical modifications of rFVIIa have been proposed, with the goal of achieving faster and longer hemostatic action. No ongoing trial is currently comparing rFVIIa variants with each other. OBJECTIVES AND METHODS We used mathematical modeling to compare the pharmacokinetics, dose-response (pharmacodynamics) and dose-effect duration (pharmacokinetics/pharmacodynamics) of rFVIIa variants to predict their optimal doses. The pharmacodynamic (PD) model of FXa generation by FVIIa in complexes with tissue factor (TF) and procoagulant lipids (PLs) was validated against published ex vivo and in vitro thrombin generation (TG) experiments. To compare variants' safety profiles, the highest non-thrombogenic doses were estimated from the clinical evidence reported for the licensed rFVIIa product. RESULTS The PD model correctly described the biphasic TF-dependent and PL-dependent dose response observed in TG experiments in vitro. The pharmacokinetic/PD simulations agreed with published ex vivo TG data for rFVIIa and the BAY 86-6150 variant, and explained the similar efficacies of a single dose of 270 μg kg(-1) (as reported in the literature) and repeated doses of 90 μg kg(-1) of unmodified rFVIIa. The duration of the simulated hemostatic effect after a single optimal dose was prolonged for rFVIIa variants with increased TF affinity or extended half-lives, but not for those with modulated PL activity. CONCLUSIONS Some modifications of the rFVIIa molecule may not translate into a prolonged hemostatic effect.
Collapse
Affiliation(s)
- A M Shibeko
- Center for Theoretical Problems of Physicochemical Pharmacology RAS, Moscow, Russia
| | | | | | | | | |
Collapse
|
15
|
Shirahata A, Fukutake K, Takamatsu J, Shima M, Hanabusa H, Mugishima H, Amano K, Takedani H, Tamashima S, Matsushita T, Tawa A, Tanaka I, Higasa S, Kosaka Y, Fujii T, Sakai M, Migita M, Kawakami K, Ohashi Y, Saito H. 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; 19:853-60. [DOI: 10.1111/hae.12205] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2013] [Indexed: 11/29/2022]
Affiliation(s)
- A. Shirahata
- Kitakyushu Yahata Higashi Hospital; Kitakyushu Fukuoka
| | - K. Fukutake
- Department of Laboratory Medicine; Tokyo Medical University; Tokyo
| | - J. Takamatsu
- Japanese Red Cross Aichi Blood Center; Seto Aichi
| | - M. Shima
- Department of Paediatrics; Nara Medical University; Kashihara Nara
| | - H. Hanabusa
- Department of Haematology; Ogikubo Hospital; Tokyo
| | - H. Mugishima
- Department of Paediatrics; Nihon University Itabashi Hospital; Tokyo
| | - K. Amano
- Department of Laboratory Medicine; Tokyo Medical University; Tokyo
| | - H. Takedani
- Department of Joint Surgery; Research Hospital of the Institute of Medical Science; The University of Tokyo; Tokyo
| | - S. Tamashima
- Department of Haematology and Oncology; Seirei Hamamatsu General Hospital; Hamamatsu Shizuoka
| | - T. Matsushita
- Department of Transfusion Medicine; Nagoya University Hospital; Nagoya
| | - A. Tawa
- Department of Paediatrics; National Hospital Organization Osaka National Hospital; Osaka
| | - I. Tanaka
- Department of Paediatrics; Nara Medical University; Kashihara Nara
| | - S. Higasa
- Division of Haematology; Department of Internal Medicine; Hyogo College of Medicine; Nishinomiya Hyogo
| | - Y. Kosaka
- Department of Haematology and Oncology; Kobe Children's Hospital; Kobe
| | - T. Fujii
- Division of the Blood Transfusion; Hiroshima University Hospital; Hiroshima
| | - M. Sakai
- Department of Paediatrics; University of Occupational and Environmental Health; Kitakyushu Fukuoka
| | - M. Migita
- Department of Paediatrics; Kumamoto Red Cross Hospital; Kumamoto
| | - K. Kawakami
- Department of Paediatrics; Kagoshima City Hospital; Kagoshima
| | - Y. Ohashi
- Department of Biostatistics; School of Public Health; The University of Tokyo; Tokyo
| | - H. Saito
- National Hospital Organization Nagoya Medical Center; Nagoya Japan
| |
Collapse
|
16
|
Tanaka KA, Kor DJ. Emerging haemostatic agents and patient blood management. Best Pract Res Clin Anaesthesiol 2013; 27:141-60. [DOI: 10.1016/j.bpa.2013.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 02/20/2013] [Indexed: 12/20/2022]
|
17
|
Shirahata A, Fukutake K, Mimaya J, Takamatsu J, Shima M, Hanabusa H, Takedani H, Takashima Y, Matsushita T, Tawa A, Higasa S, Takata N, Sakai M, Kawakami K, Ohashi Y, Saito H. Results of clot waveform analysis and thrombin generation test for a plasma-derived factor VIIa and X mixture (MC710) in haemophilia patients with inhibitors-phase I trial: 2nd report. Haemophilia 2012; 19:330-7. [DOI: 10.1111/hae.12024] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2012] [Indexed: 11/27/2022]
Affiliation(s)
- A. Shirahata
- Department of Paediatrics; University of Occupational and Environmental Health; Kitakyushu; Fukuoka; Japan
| | - K. Fukutake
- Department of Laboratory Medicine; Tokyo Medical University; Tokyo; Japan
| | - J. Mimaya
- Division of Haematology and Oncology; Shizuoka Children's Hospital; Shizuoka; Japan
| | - J. Takamatsu
- Department of Transfusion Medicine; Nagoya University Hospital; Nagoya; Japan
| | - M. Shima
- Department of Paediatrics; Nara Medical University; Kashihara; Nara; Japan
| | - H. Hanabusa
- Department of Haematology; Ogikubo Hospital; Tokyo; Japan
| | - H. Takedani
- Department of Joint Surgery, Research Hospital of the Institute of Medical Science; the University of Tokyo; Tokyo; Japan
| | - Y. Takashima
- Division of Haematology and Oncology; Shizuoka Children's Hospital; Shizuoka; Japan
| | - T. Matsushita
- Department of Haematology and Oncology; Nagoya University Graduate School of Medicine; Nagoya; Japan
| | - A. Tawa
- Department of Paediatrics; National Hospital Organization Osaka National Hospital; Osaka; Japan
| | - S. Higasa
- Division of Haematology, Department of Internal Medicine; Hyogo College of Medicine; Nishinomiya; Hyogo; Japan
| | - N. Takata
- Division of the Blood Transfusion Services; Hiroshima University Hospital; Hiroshima; Japan
| | - M. Sakai
- Department of Paediatrics; University of Occupational and Environmental Health; Kitakyushu; Fukuoka; Japan
| | - K. Kawakami
- Department of Paediatrics; Kagoshima City Hospital; Kagoshima; Japan
| | - Y. Ohashi
- Department of Biostatistics, School of Public Health; the University of Tokyo; Tokyo; Japan
| | - H. Saito
- Nagoya Central Hospital; Nagoya; Japan
| |
Collapse
|
18
|
Nakatomi Y, Tsuji M, Gokudan S, Hanada-Dateki T, Nakashima T, Miyazaki H, Hamamoto T, Nakagaki T, Tomokiyo K. Stable complex formation between serine protease inhibitor and zymogen: coagulation factor X cleaves the Arg393-Ser394 bond in a reactive centre loop of antithrombin in the presence of heparin. J Biochem 2012; 152:463-70. [PMID: 22923734 DOI: 10.1093/jb/mvs094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Antithrombin (AT) inhibits several blood coagulation proteases, including activated factor X (FXa), by forming stable complexes with these proteases. Herein, we demonstrate that AT forms a stable complex with zymogen factor X (FX). Sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) and size-exclusion chromatography analyses showed that AT and FX formed an SDS-stable complex, which is distinct in apparent molecular mass from an FXa-AT complex, in the presence of heparin. Amino-terminal sequence analysis of the complex following SDS-PAGE under reducing conditions provided clear evidence that AT forms this complex with the heavy chain of FX, because two sequences, HGSPVDI (residues 1-7 of AT) and SVAQATS (residues 1-7 of the heavy chain of FX), were identified. Furthermore, sequence SLNPNRV, which corresponds to residues 394-400 of AT, was identified in the non-reduced FX-AT complex, indicating that FX cleaved the Arg393-Ser394 bond in a reactive centre loop of AT. Unfractionated heparin induced FX-AT complex formation more effectively than low-molecular weight heparin or AT-binding pentasaccharide, and appeared to promote complex formation mainly via a template effect. These data suggest that AT is capable of forming a stable complex with zymogen FX by acting as an inhibitor in the presence of heparin.
Collapse
Affiliation(s)
- Yasushi Nakatomi
- Therapeutic Protein Product Research Department, The Chemo-Sero-Therapeutic Research Institute, KAKETSUKEN, 1-6-1 Okubo, Kumamoto-shi, Kumamoto 860-8568, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Pharmacokinetics, distribution, and excretion of 125I-labeled human plasma-derived-FVIIa and -FX with MC710 (FVIIa/FX mixture) in rats. Thromb Res 2012; 129:62-7. [DOI: 10.1016/j.thromres.2011.04.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 03/16/2011] [Accepted: 04/26/2011] [Indexed: 11/19/2022]
|
20
|
Ingerslev J, Sørensen B. Parallel use of by-passing agents in haemophilia with inhibitors: a critical review. Br J Haematol 2011; 155:256-62. [DOI: 10.1111/j.1365-2141.2011.08854.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
21
|
SHIRAHATA A, FUKUTAKE K, MIMAYA J, TAKAMATSU J, SHIMA M, HANABUSA H, TAKEDANI H, TAKASHIMA Y, MATSUSHITA T, TAWA A, HIGASA S, TAKATA N, SAKAI M, KAWAKAMI K, OHASHI Y, SAITO H. Clinical pharmacological study of a plasma-derived factor VIIa and factor X mixture (MC710) in haemophilia patients with inhibitors - Phase I trial. Haemophilia 2011; 18:94-101. [DOI: 10.1111/j.1365-2516.2011.02548.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
22
|
Nishiura H, Tanase S, Tsujita K, Sugiyama S, Ogawa H, Nakagaki T, Semba U, Yamamoto T. Maintenance of ribosomal protein S19 in plasma by complex formation with prothrombin. Eur J Haematol 2011; 86:436-41. [DOI: 10.1111/j.1600-0609.2011.01585.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
23
|
Future pharmacological strategies in management of Haemophilia. Thromb Res 2010; 126:259-61. [DOI: 10.1016/j.thromres.2010.01.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Revised: 01/18/2010] [Accepted: 01/20/2010] [Indexed: 11/22/2022]
|