Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Haemophilia ; 29(6): 1633-1637, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37824563

RESUMEN

INTRODUCTION AND AIMS: Mim8 is a next generation bispecific antibody developed for the treatment of haemophilia A (HA). Mim8 has an increased potency compared to first generation molecules. The impact on Mim8 on non-FVIII measuring haemostasis assays was assessed in plasma containing Mim8. METHODS: Congenital severe HA plasma was spiked with increasing concentrations of Mim8 (0-20 µg/mL). 28 routine and specialist haemostasis assays were used to measure activities. These included tests for prothrombin time (PT), fibrinogen, thrombin, D-dimer, anti-Xa, heparin induced thrombocytopenia (HIT), clotting factors II-XII, factor XIII, von Willebrand factor (VWF), thrombophilia and DRVVT. RESULTS AND CONCLUSIONS: Less than 10 % difference was calculated between plasma without Mim8 and plasma spiked to 15 µg/mL Mim8 in all assays except thrombin time (-10.5%), APTT-based factor IX, XI and XII, Werfen VWF:RCo (10.6%) and Siemens LA1 (-26.4%) and LA2 (-16.9%). At the expected therapeutic steady state levels of Mim8 (5-8 µg/mL), less than 10% difference was calculated for thrombin time and Werfen VWF:RCo. APTT-based assays of FIX, XI and XII are significantly elevated in the presence of Mim8 and should not be performed. A chromogenic FIX assay could be used to accurately measure FIX activity in the presence of Mim8. There was some interference in the DRVVT method we used so local assessment of other DRVVT methods is advised. Differences in all other tests would not be predicted to affect patient management.


Asunto(s)
Coagulación Sanguínea , Hemofilia A , Humanos , Factor de von Willebrand/uso terapéutico , Hemostasis , Pruebas de Coagulación Sanguínea/métodos , Tiempo de Protrombina , Hemofilia A/tratamiento farmacológico
2.
Haemophilia ; 27(3): e389-e392, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33314455

RESUMEN

FVIII activity in samples taken at various time points from 21 patients treated with N8-GP (Esperoct® ; turoctocog alfa pegol) during the pathfinder clinical trial programme was assessed and compared using different assay methods. FVIII activity measurements in samples from patients treated with N8-GP were similar using chromogenic assays, regardless of calibration method or kit/analyser combination. FVIII activity measurements using one-stage aPTT-based assays were slightly lower when calibrated using normal human plasma (NHP) compared with a product-specific standard; this difference may be partially attributable to differences in the aPTT reagent/analyser combinations used to perform the measurements. Overall, these results confirm the accuracy of FVIII activity measurements using N8-GP-treated patient samples and routine clinical laboratory methods with NHP calibration.


Asunto(s)
Factor VIII , Hemofilia A , Pruebas de Coagulación Sanguínea , Hemofilia A/tratamiento farmacológico , Humanos , Tiempo de Tromboplastina Parcial
3.
Haemophilia ; 26(1): 156-163, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31809565

RESUMEN

Factor replacement therapy with factor VIII (FVIII) concentrates is the current standard of care for patients with haemophilia A. Postadministration monitoring of FVIII activity during on-demand or prophylactic treatment is important, for example to guide a suitable dosing regimen. While the use of two-stage chromogenic substrate (CS) assays is increasing, activated partial thromboplastin time (APTT)-based one-stage clotting (OSC) assays are most commonly used to measure FVIII activity in clinical laboratories. Substantial variations in activity measurements have been observed in association with some OSC assay reagents when assessing extended half-life FVIII molecules. Certain silica-based APTT reagents have previously been shown to underestimate FVIII activity with the polyethylene glycol (PEG)-conjugated product turoctocog alfa pegol (N8-GP [ESPEROCT® ]; Novo Nordisk A/S). As a wide range of assay reagents are used in clinical laboratories worldwide, it is essential to establish which can be used to accurately measure activity with modified FVIII concentrates. Here, we describe the approach taken by Novo Nordisk to determine the suitability and accuracy of assays and reagents to measure FVIII activity in samples that contain N8-GP. While accurate activity measurements were possible with all tested CS assays and most of the OSC APTT reagents tested, three APTT reagents that contain silica as a contact activator were found to underestimate N8-GP recovery (APTT-SP, TriniCLOT™, STA® PTT-Automate). The data demonstrate the importance of characterizing the accuracy of each FVIII activity assay. Any limitations should be communicated to treating physicians and the clinical laboratories that test samples containing N8-GP.


Asunto(s)
Bioensayo , Monitoreo de Drogas , Factor VIII/administración & dosificación , Factor VIII/uso terapéutico , Polietilenglicoles/química , Glicosilación , Humanos , Tiempo de Tromboplastina Parcial
4.
Acta Haematol ; 143(5): 504-508, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31639805

RESUMEN

N8-GP (ESPEROCT®; turoctocog alfa pegol; Novo Nordisk A/S, Bagsvaerd, Denmark) is an extended half-life recombinant factor VIII (FVIII) molecule. FVIII-deficient plasma spiked with N8-GP can be accurately measured using many activated partial thromboplastin time (aPTT)-based one-stage clotting assay reagents with normal human plasma calibrators. To date, there are few data on the measurement accuracy of samples from patients treated with N8-GP. Here, we measure patient samples during routine treatment monitoring. Three previously treated patients with severe hemophilia A (HA) without inhibitors (baseline FVIII activity <0.01 IU/mL) received 50 IU/kg N8-GP every fourth day or twice weekly over 5 years as part of the pathfinder2 trial. Patient samples were monitored using the Pathromtin® SL aPTT reagent (Siemens Healthcare GmbH, Erlangen, Germany), a BCS® XP System analyzer (Siemens), and Standard Human Plasma (Siemens) or product-specific calibrators. Patient age ranged from 36 to 62 years. Overall, measurements performed using product-specific or Standard Human Plasma calibrators were in good agreement, with ratios randomly distributed around 1.0. Peak ratios tended to be closer to 1.0 than trough samples. Pathromtin® SL with Standard Human Plasma calibrator consistently and accurately measured FVIII activity in samples from severe HA patients receiving N8-GP prophylaxis.


Asunto(s)
Pruebas de Coagulación Sanguínea/métodos , Factor VIII/análisis , Hemofilia A/patología , Adulto , Factor VIII/uso terapéutico , Hemofilia A/sangre , Hemofilia A/tratamiento farmacológico , Humanos , Masculino , Persona de Mediana Edad
5.
Blood ; 129(15): 2161-2171, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28039188

RESUMEN

Wound healing requires interactions between coagulation, inflammation, angiogenesis, cellular migration, and proliferation. Healing in dermal wounds of hemophilia B mice is delayed when compared with hemostatically normal wild-type (WT) mice, with abnormal persistence of iron deposition, inflammation, and neovascularity. We observed healing following induced joint hemorrhage in WT and factor IX (FIX) knockout (FIX-/-) mice, examining also parameters previously studied in an excisional skin wound model. Hemostatically normal mice tolerated this joint bleeding challenge, cleared blood from the joint, and healed with minimal pathology, even if additional autologous blood was injected intra-articularly at the time of wounding. Following hemarthrosis, joint wound healing in hemophilia B mice was impaired and demonstrated similar abnormal histologic features as previously described in hemophilic dermal wounds. Therefore, studies of pathophysiology and therapy of hemophilic joint bleeding performed in hemostatically normal animals are not likely to accurately reflect the healing defect of hemophilia. We additionally explored the hypothesis that the use of a FIX replacement protein with extended circulating FIX activity could improve synovial and osteochondral wound healing in hemophilic mice, when compared with treatment with unmodified recombinant FIX (rFIX) in the established joint bleeding model. Significantly improved synovial wound healing and preservation of normal osteochondral architecture are achieved by extending FIX activity after hemarthrosis using glycoPEGylated FIX when compared with an equivalent dose of rFIX. These results suggest that treating joint bleeding only until hemostasis is achieved may not result in optimal joint healing, which is improved by extending factor activity.


Asunto(s)
Factor IX , Hemartrosis , Hemofilia B , Articulaciones , Piel , Cicatrización de Heridas , Animales , Modelos Animales de Enfermedad , Factor IX/genética , Factor IX/farmacología , Hemartrosis/tratamiento farmacológico , Hemartrosis/genética , Hemartrosis/metabolismo , Hemofilia B/tratamiento farmacológico , Hemofilia B/genética , Hemofilia B/metabolismo , Articulaciones/lesiones , Articulaciones/metabolismo , Ratones , Ratones Noqueados , Piel/lesiones , Piel/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética
6.
Haemophilia ; 25(1): 154-161, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30664825

RESUMEN

N9-GP (nonacog beta pegol; Refixia® ; Rebinyn® , Novo Nordisk A/S, Bagsvaerd, Denmark) is a glycoPEGylated extended half-life recombinant factor IX (rFIX) that exhibits efficacy and potency comparable to unmodified FIX molecules in non-clinical models. Phase 3 clinical trials have confirmed the efficacy and tolerability of N9-GP for the prevention and on-demand treatment of bleeding episodes in patients with haemophilia B. Recent studies have shown that PEGylation affects clotting times in activated partial thromboplastin time (aPTT)-based one-stage activity assays due to interaction between the FIX molecule and certain aPTT reagents. In recognition of the challenges surrounding FIX activity assessment, the identification of consistent, reproducible and accurate assays to measure FIX activity has been a priority for Novo Nordisk, running in parallel to the clinical development program for N9-GP. N9-GP activity can be reliably measured using chromogenic substrate assays and specific aPTT reagents. The conjugation of the PEG moiety to the FIX molecule may affect one-stage aPTT-based clotting assays in a reagent-dependent manner. Many aPTT reagents that use silica as the contact activator dramatically overestimate N9-GP activity due to premature activation. On the other hand, the contact activator in some other aPTT reagents negatively affects the enzymatic activity of FXIa, causing the underestimation of N9-GP activity. While N9-GP activity cannot be measured consistently with all available aPTT reagents, accurate N9-GP measurements can be achieved with certain aPTT reagents. Here, we review the studies that led to these findings and summarize the current options for accurate measurement of N9-GP in patient samples.


Asunto(s)
Pruebas de Coagulación Sanguínea/métodos , Factor IX/análisis , Polietilenglicoles/análisis , Monitoreo de Drogas , Factor IX/uso terapéutico , Hemofilia B/tratamiento farmacológico , Humanos , Tiempo de Tromboplastina Parcial , Polietilenglicoles/uso terapéutico , Proteínas Recombinantes/análisis , Proteínas Recombinantes/uso terapéutico
7.
Haemophilia ; 25(5): 893-901, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31294905

RESUMEN

AIM: N8-GP (turoctocog alfa pegol) is a glycoPEGylated, extended half-life human recombinant factor VIII (FVIII) shown to be an efficacious treatment for patients with haemophilia A. Accurate monitoring of replacement therapy helps ensure proper dosing, leading to better patient care. The objective of this field study was to evaluate the accuracy and intra- and inter-laboratory variabilities of N8-GP and rAHF (Advate® ) FVIII activity (FVIII:C) measurements in clinical laboratories using their routine methods and reagents. METHODS: Laboratories measured plasma samples spiked with 0.03, 0.2, 0.6 and 0.9 IU/mL N8-GP or rAHF. Samples were blinded, and laboratories were instructed to perform evaluations using their routine FVIII activity assays and calibrators. RESULTS: Of the 67 participating laboratories from 25 countries, 60 used a one-stage assay, 36 used a chromogenic assay, and 29 used both one-stage and chromogenic assays. Participating laboratories used nine different activated partial thromboplastin time (aPTT) reagents, the most common being SynthASil® and Actin® FS. Most aPTT reagents recovered N8-GP close to target. Three silica-based aPTT reagents (APTT-SP, TriniCLOT™ and STA® PTT-Automate) underestimated N8-GP, recovering 40%-83% of target concentration. For chromogenic assays, N8-GP and rAHF recoveries were comparable at all concentrations, with overall mean recoveries for both products close to 130%. Assay variability was similar for both assay types and both products; inter-laboratory variability was greater than intra-laboratory variability and highest at 0.03 IU/mL. CONCLUSIONS: Most clinical laboratories accurately measured N8-GP and rAHF when using their in-house one-stage or chromogenic FVIII:C assays. However, three silica-based aPTT reagents underestimated N8-GP recovery.


Asunto(s)
Factor VIII/metabolismo , Hemostasis/fisiología , Pruebas de Coagulación Sanguínea/métodos , Humanos , Laboratorios
8.
Haemophilia ; 24(5): 815-822, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30112856

RESUMEN

INTRODUCTION: The thrombin generation assay (TGA) can be used to monitor factor replacement therapy in patients with haemophilia. The TGA assay is typically performed using tissue factor as the reaction activator; however, activating with FIXa or FXIa can enhance assay sensitivity when FVIII < 1%. AIMS: To evaluate the sensitivity of the TGA when FIXa (5 nmol/L) and FXIa (0.22 nmol/L) are used to activate the assay in platelet-poor plasma and to compare these data to the one-stage and chromogenic assays. METHODS: Plasma from 10 severe FVIII-deficient subjects was supplemented with FVIII (0%, 0.1%, 0.4%, 1.2%, 4%, 11% and 33%), using either Novo Eight® , Advate® , Eloctate® , turoctocog alfa pegol or a control standard. The one-stage and chromogenic assays quantified the FVIII levels. The TGA assay was activated using either FIXa or FXIa. RESULTS: Both FIXa- and FXIa-activated TGA were sensitive across FVIII concentrations, with intra-assay coefficient of variation (CV) < 10%. The FXIa-activated assay had 25% CV at the lowest level of FVIII compared to 10% CV with FIXa activation. There were strong correlations between the FIXa- and FXIa-activated TGA tests (R2  = 0.9912) and between the one-stage and chromogenic assays (R2  = 0.9469). However, there were poor relationships between the TGA tests and one-stage and chromogenic assays. CONCLUSIONS: Both FIXa- and FXIa activation results in similar TGA profiles across a FVIII range of 0.1%-33%; however, FIXa activation was more robust at the lowest levels of FVIII compared with FXIa activation.


Asunto(s)
Factor VIII/metabolismo , Factor XIa/metabolismo , Hemofilia A/sangre , Trombina/metabolismo , Humanos
9.
Blood ; 121(11): 2108-16, 2013 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-23335368

RESUMEN

Frequent infusions of intravenous factor VIII (FVIII) are required to prevent bleeding associated with hemophilia A. To reduce the treatment burden, recombinant FVIII with a longer half-life was developed without changing the protein structure. FVIII-polyethylene glycol (PEG) conjugates were prepared using an enzymatic process coupling PEG (ranging from 10 to 80 kDa) selectively to a unique O-linked glycan in the FVIII B-domain. Binding to von Willebrand factor (VWF) was maintained for all conjugates. Upon cleavage by thrombin, the B-domain and the associated PEG were released, generating activated FVIII (FVIIIa) with the same primary structure and specific activity as native FVIIIa. In both FVIII- and VWF-deficient mice, the half-life was found to increase with the size of PEG. In vivo potency and efficacy of FVIII conjugated with a 40-kDa PEG (N8-GP) and unmodified FVIII were not different. N8-GP had a longer duration of effect in FVIII-deficient mouse models, approximately a twofold prolonged half-life in mice, rabbits, and cynomolgus monkeys; however, the prolongation was less pronounced in rats. Binding capacity of N8-GP on human monocyte-derived dendritic cells was reduced compared with unmodified FVIII, resulting in several-fold reduced cellular uptake. In conclusion, N8-GP has the potential to offer efficacious prevention and treatment of bleeds in hemophilia A at reduced dosing frequency.


Asunto(s)
Factor VIII/uso terapéutico , Hemofilia A/tratamiento farmacológico , Polietilenglicoles/uso terapéutico , Animales , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Factor VIII/administración & dosificación , Factor VIII/metabolismo , Femenino , Glicosilación , Hemofilia A/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Polietilenglicoles/administración & dosificación , Polietilenglicoles/metabolismo , Conejos , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Resultado del Tratamiento
10.
Blood ; 119(24): 5871-8, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22563084

RESUMEN

Hemophilia is treated by IV replacement therapy with Factor VIII (FVIII) or Factor IX (FIX), either on demand to resolve bleeding, or as prophylaxis. Improved treatment may be provided by drugs designed for subcutaneous and less frequent administration with a reduced risk of inhibitor formation. Tissue factor pathway inhibitor (TFPI) down-regulates the initiation of coagulation by inhibition of Factor VIIa (FVIIa)/tissue factor/Factor Xa (FVIIa/TF/FXa). Blockage of TFPI inhibition may facilitate thrombin generation in a hemophilic setting. A high-affinity (K(D) = 25pM) mAb, mAb 2021, against TFPI was investigated. Binding of mAb 2021 to TFPI effectively prevented inhibition of FVIIa/TF/FXa and improved clot formation in hemophilia blood and plasma. The binding epitope on the Kunitz-type protease inhibitor domain 2 of TFPI was mapped by crystallography, and showed an extensive overlap with the FXa contact region highlighting a structural basis for its mechanism of action. In a rabbit hemophilia model, an intravenous or subcutaneous dose significantly reduced cuticle bleeding. mAb 2021 showed an effect comparable with that of rFVIIa. Cuticle bleeding in the model was reduced for at least 7 days by a single intravenous dose of mAb 2021. This study suggests that neutralization of TFPI by mAb 2021 may constitute a novel treatment option in hemophilia.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor Xa/metabolismo , Hemofilia A/tratamiento farmacológico , Hemostasis/efectos de los fármacos , Lipoproteínas/metabolismo , Modelos Moleculares , Animales , Anticuerpos Bloqueadores/administración & dosificación , Anticuerpos Bloqueadores/uso terapéutico , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Neutralizantes/farmacología , Tiempo de Sangría , Coagulación Sanguínea/efectos de los fármacos , Reacciones Cruzadas/efectos de los fármacos , Modelos Animales de Enfermedad , Epítopos/inmunología , Factor VIII/farmacología , Factor Xa/inmunología , Femenino , Fibrina/metabolismo , Células HEK293 , Hemofilia A/sangre , Células Endoteliales de la Vena Umbilical Humana , Humanos , Pruebas de Neutralización , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Conejos , Especificidad de la Especie , Tromboplastina/farmacología
11.
Eur J Haematol ; 93(5): 369-76, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24797664

RESUMEN

Turoctocog alfa (NovoEight®) is a recombinant factor VIII (rFVIII) with a truncated B-domain made from the sequence coding for 10 amino acids from the N-terminus and 11 amino acids from the C-terminus of the naturally occurring B-domain. Turoctocog alfa is produced in Chinese hamster ovary (CHO) cells without addition of any human- or animal-derived materials. During secretion, some rFVIII molecules are cleaved at the C-terminal of the heavy chain (HC) at amino acid 720, and a monoclonal antibody binding C-terminal to this position is used in the purification process allowing isolation of the intact rFVIII. Viral inactivation is ensured by a detergent inactivation step as well as a 20-nm nano-filtration step. Characterisation of the purified protein demonstrated that turoctocog alfa was fully sulphated at Tyr346 and Tyr1664, which is required for optimal proteolytic activation by thrombin. Kinetic assessments confirmed that turoctocog alfa was activated by thrombin at a similar rate as seen for other rFVIII products fully sulphated at these positions. Tyr1680 was also fully sulphated in turoctocog alfa resulting in strong affinity (low nm Kd ) for binding to von Willebrand factor (VWF). Half-lives of 7.2 ± 0.9 h in F8-KO mice and 8.9 ± 1.8 h haemophilia A dogs supported that turoctocog alfa bound to VWF after infusion. Functional studies including thromboelastography analysis of human haemophilia A whole blood with added turoctocog alfa and effect studies in mice bleeding models demonstrated a dose-dependent effect of turoctocog alfa. The non-clinical data thus confirm the haemostatic effect of turoctocog alfa and, together with the comprehensive clinical evaluation, support the use as FVIII replacement therapy in patients with haemophilia A.


Asunto(s)
Factor VIII/farmacocinética , Procesamiento Proteico-Postraduccional , Animales , Células CHO , Cricetulus , Perros , Evaluación Preclínica de Medicamentos , Factor VIII/biosíntesis , Factor VIII/genética , Factor VIII/aislamiento & purificación , Expresión Génica , Glicosilación , Semivida , Hemofilia A/tratamiento farmacológico , Humanos , Cinética , Ratones , Modificación Traduccional de las Proteínas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacocinética , Sulfatos , Trombina/metabolismo
12.
Blood ; 118(8): 2333-41, 2011 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-21700771

RESUMEN

Current management of hemophilia B entails multiple weekly infusions of factor IX (FIX) to prevent bleeding episodes. In an attempt to make a longer acting recombinant FIX (rFIX), we have explored a new releasable protraction concept using the native N-glycans in the activation peptide as sites for attachment of polyethylene glycol (PEG). Release of the activation peptide by physiologic activators converted glycoPEGylated rFIX (N9-GP) to native rFIXa and proceeded with normal kinetics for FXIa, while the K(m) for activation by FVIIa-tissue factor (TF) was increased by 2-fold. Consistent with minimal perturbation of rFIX by the attached PEG, N9-GP retained 73%-100% specific activity in plasma and whole-blood-based assays and showed efficacy comparable with rFIX in stopping acute bleeds in hemophilia B mice. In animal models N9-GP exhibited up to 2-fold increased in vivo recovery and a markedly prolonged half-life in mini-pig (76 hours) and hemophilia B dog (113 hours) compared with rFIX (16 hours). The extended circulation time of N9-GP was reflected in prolonged correction of coagulation parameters in hemophilia B dog and duration of effect in hemophilia B mice. Collectively, these results suggest that N9-GP has the potential to offer efficacious prophylactic and acute treatment of hemophilia B patients at a reduced dosing frequency.


Asunto(s)
Factor IX/química , Factor IX/metabolismo , Animales , Sitios de Unión , Modelos Animales de Enfermedad , Perros , Factor IX/genética , Femenino , Semivida , Hemofilia B/sangre , Hemofilia B/tratamiento farmacológico , Hemofilia B/genética , Hemostáticos/sangre , Hemostáticos/química , Hemostáticos/farmacología , Humanos , Técnicas In Vitro , Cinética , Masculino , Ratones , Ratones Mutantes , Polietilenglicoles/química , Polisacáridos/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Porcinos , Porcinos Enanos
13.
J Thromb Haemost ; 21(6): 1493-1502, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37037699

RESUMEN

BACKGROUND: Mim8 is a novel antifactor IXa/antifactor X bispecific antibody in clinical development for prophylactic treatment of hemophilia A with and without inhibitors. Patients treated with Mim8 may need supplementary bleed treatment under certain conditions such as surgery or major trauma. OBJECTIVES: This study aimed to better understand the response of Mim8 in thrombin generation assays (TGAs) alone or in combination with other hemostatic proteins. METHODS: We used TGAs with different activators (tissue factor (TF) and activated factor XI) to better understand the similarities and differences between the mode of action of Mim8 and factor VIII (FVIII). Following this, we investigated the effects of mixing Mim8 with the main bleed treatment options for persons with hemophilia A with or without inhibitors: FVIII, activated factor VII (FVIIa), and activated prothrombin complex concentrates (aPCC). RESULTS: The results indicated that for patients without inhibitors, Mim8 does not interfere with FVIII's mode of action. For patients with inhibitors, Mim8 mixed with aPCC results in a strong synergistic effect causing thrombin generation far exceeding the normal levels. Contrary to this, mixing Mim8 with FVIIa results in a more controlled additive effect, visible only when using TF as a trigger, which does not exceed the normal level of thrombin generation. CONCLUSION: These findings support the use of approved clinical doses of FVIIa for bleed treatment of patients with FVIII inhibitors treated with Mim8. Additionally, the findings suggest that concomitant use of FVIII and Mim8 is safe for managing breakthrough bleeds.


Asunto(s)
Hemofilia A , Hemostáticos , Humanos , Factor VIII , Trombina/metabolismo , Factor VIIa/metabolismo , Hemostáticos/uso terapéutico , Hemorragia/tratamiento farmacológico , Factor IX/uso terapéutico , Tromboplastina/uso terapéutico , Proteínas Recombinantes/uso terapéutico
14.
J Thromb Haemost ; 21(3): 480-487, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36696203

RESUMEN

BACKGROUND: Mim8 is a next generation bispecific antibody developed for the prophylactic treatment of hemophilia A. The sensitivity of activated plasma thromboplastin time (APTT), assays measuring factor VIII activity (FVIII:C) and thrombin generation to plasma containing Mim8 was assessed. METHODS: Congenital severe hemophilia A plasma was spiked with Mim8 at 0 µg/mL to 20 µg/mL. APTT was measured with 4 reagents, one-stage FVIII with 9 reagents, and chromogenic FVIII with 6 assays. Thrombin generation was assessed in a low tissue factor system. RESULTS: At 1-µg/mL Mim8, the APTT was shortened to within normal limits and one-stage FVIII:C was >1.00 IU/mL with all APTT reagents. Modified one-stage assays calibrated with Mim8 reference material calibrated recovered within 20% of the target Mim8 concentration with most APTT reagents. Factor VIII:C of bovine only chromogenic assays was <0.04 IU/mL at all Mim8 concentrations. Factor VIII:C of human only chromogenic assay was >4.00 IU/mL at 20-µg/mL Mim8. Factor VIII:C of hybrid bovine FX, human FIXa chromogenic assays ranged from 0.076 to >3.00 IU/mL at 20-µg/mL Mim8. Normal thrombin generation was restored at 5-µg/mL Mim8. CONCLUSIONS: APTT-based assays are sensitive to Mim8 and should not be performed in the presence of the drug. Chromogenic assays containing human proteins or hybrid human/bovine proteins demonstrated variable sensitivity to Mim8. Bovine only chromogenic assays were largely insensitive to the presence of Mim8. Thrombin generation normalized at increased Mim8 concentrations. Modified one-stage and chromogenic assays could be used to quantify the Mim8 concentration in plasma.


Asunto(s)
Anticuerpos Biespecíficos , Hemofilia A , Hemostáticos , Animales , Bovinos , Humanos , Factor VIII/metabolismo , Hemofilia A/tratamiento farmacológico , Trombina/metabolismo , Tiempo de Tromboplastina Parcial , Hemostáticos/uso terapéutico , Tromboplastina/uso terapéutico , Indicadores y Reactivos , Anticuerpos Biespecíficos/uso terapéutico
15.
Semin Thromb Hemost ; 38(3): 274-81, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22510860

RESUMEN

Vatreptacog alfa is a genetically engineered variant of recombinant factor VIIa (rFVIIa) containing three amino acid changes. Aspartic acid, valine, and glutamine residues replace valine, glutamic acid, and methionine at positions 158, 296, and 298, respectively. These substitutions result in considerable enhancement of the intrinsic (tissue factor-independent) capability to activate factor X and the downstream hemostatic events are consequently augmented. The beneficial effects of vatreptacog alfa have been demonstrated in numerous in vitro systems attempting to mimic hemophilia and corroborated in in vivo models. Vatreptacog alfa has successfully passed through phase 1 and 2 clinical trials and the molecule is currently being explored in phase 3 clinical trial for the treatment of bleedings in hemophilia patients with inhibitors. This article describes the proposed mechanism behind the increased activity and action of vatreptacog alfa and reviews available data, which suggest that vatreptacog alfa could be a valuable addition to the existing portfolio of treatment options for hemophilia patients with inhibitors.


Asunto(s)
Factor VIIa/farmacología , Hemofilia A/tratamiento farmacológico , Animales , Células CHO , Cricetinae , Factor VIIa/genética , Ingeniería Genética , Humanos , Estructura Molecular , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
16.
Br J Haematol ; 152(1): 99-107, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21083658

RESUMEN

NN1731 is a recombinant activated factor VII (rFVIIa) analogue with increased intrinsic activity. This also applies to its reactivity towards antithrombin (AT), the role of which was investigated in a pharmacokinetic (PK) study. NN1731 or rFVIIa was administered to normal and haemophilia A dogs and elimination was measured by FVIIa clot activity, FVIIa- and FVIIa-AT antigen. In vitro AT complex formation was studied in canine plasma spiked with NN1731 or rFVIIa. Based on FVIIa antigen concentrations, PK profiles in normal and haemophilia A dogs were similar for NN1731 and rFVIIa with antigen half lives, t(½) ≈1·8 h. In contrast, PK profiles based on activity measurements were distinctly different. NN1731 induced a strong, short lasting (t(½) ≈0·5 h) pro-coagulant response, whereas rFVIIa induced a lower, longer lasting (t(½) ≈1·1 h) response. Western Blot and FVIIa-AT antigen analysis demonstrated in vivo AT complex formation that accounted for these divergences. AT complex formation with FVIIa or NN1731 in vitro in canine plasma was considerably slower than the in vivo reaction. The results suggest that in vivo inhibition by AT contributes significantly to define drug duration in haemophilia treatment with rFVIIa and in particular with the NN1731 analogue.


Asunto(s)
Proteínas Antitrombina/fisiología , Coagulantes/farmacocinética , Factor VII/farmacocinética , Hemofilia A/sangre , Animales , Coagulación Sanguínea/efectos de los fármacos , Inhibidores de Factor de Coagulación Sanguínea/fisiología , Coagulantes/antagonistas & inhibidores , Modelos Animales de Enfermedad , Perros , Factor VII/antagonistas & inhibidores , Factor VIIa/antagonistas & inhibidores , Semivida , Masculino , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/sangre , Tromboelastografía/métodos
17.
Blood ; 113(16): 3682-9, 2009 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-19109232

RESUMEN

Continuous expression of activated factor VII (FVIIa) via gene transfer is a potential therapeutic approach for hemophilia patients with or without inhibitory antibodies to human factor VIII (FVIII) or IX (FIX). Here, we investigate whether gene transfer of an engineered canine FVIIa (cFVIIa) transgene can affect hemostasis in a canine model of hemophilia, a good predictor of efficacy of hemophilia treatments. Purified recombinant cFVIIa exhibited 12-fold higher tissue factor-dependent activity than purified recombinant zymogen cFVII. Subsequently, we generated a serotype 8 recombinant adeno-associated viral vector expressing cFVIIa from a liver-specific promoter. Vector delivery via the portal vein in hemophilia A and B dogs was well tolerated, and long-term expression of cFVIIa resulted in a shortening of the prothrombin time, partial correction of the whole blood clotting time and thromboelastography parameters, and a complete absence of spontaneous bleeding episodes. No evidence of hepatotoxicity, thrombotic complications, or inhibitory immune response was found. These data provide the first evidence for in vivo efficacy and safety of continuously expressed FVIIa as a FVIII/FIX-bypassing agent in a large animal model of hemophilia, avoiding the risk of inhibitor formation associated with bolus FVIII or FIX infusion.


Asunto(s)
Dependovirus , Expresión Génica , Terapia Genética , Hemofilia A/terapia , Animales , Inhibidores de Factor de Coagulación Sanguínea/genética , Inhibidores de Factor de Coagulación Sanguínea/metabolismo , Modelos Animales de Enfermedad , Perros , Factor VIIa , Hemofilia A/genética , Hemofilia A/metabolismo , Hemorragia/genética , Hemorragia/metabolismo , Hemorragia/terapia , Humanos , Hígado/metabolismo , Especificidad de Órganos , Regiones Promotoras Genéticas , Tiempo de Protrombina , Tromboelastografía
18.
J Trauma ; 69(5): 1196-202, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20032791

RESUMEN

BACKGROUND: Colloid plasma expanders are used to maintain blood pressure and ensure tissue perfusion during hypovolemia, e.g., caused by traumatic bleeding. Although colloids stabilize the cardiovascular system, they can also potentially cause coagulopathy. Consequently, bleeding tendency may increase, as well as the associated risk of morbidity and mortality. Thus, there is a need for hemostatic treatment options for these patients. rFVIIa (NovoSeven, Novo Nordisk A/S, Bagsvaerd, Denmark) is a hemostatic agent that effectively controls bleedings in patients with inhibitor-complicated hemophilia. rFVIIa works by enhancing thrombin generation on the activated platelet surface at the site of injury, leading to the formation of a stable fibrin clot. NN1731 is an rFVIIa analog with increased hemostatic potential and is currently under clinical development. METHODS: In this study, the effect of rFVIIa and NN1731 on cuticle bleeding in rabbits 50% hemodiluted with hydroxyethyl starch (molecular weight ∼ 200,000) was tested. Cuticle bleeding was induced after a two-stage hemodilution procedure. After 5 minutes, the animals were treated with rFVIIa (2, 5, or 10 mg/kg), NN1731 (1 or 2 mg/kg), or vehicle, followed by 30 minutes of observation. RESULTS: Hemodilution caused a significant increase in bleeding time and blood loss. rFVIIa dose-dependently reduced bleeding time and blood loss, reaching statistical significance at 10 mg/kg. However, 2 mg/kg NN1731 reduced bleeding time and blood loss significantly and to a similar extent as 10 mg/kg rFVIIa. This increased hemostatic potential of NN1731 compared with rFVIIa and was confirmed by findings using thromboelastography on ex vivo hemodiluted whole blood. CONCLUSION: In summary, rFVIIa and NN1731 significantly and dose-dependently reduced bleeding in extensively hemodiluted rabbits.


Asunto(s)
Factor VII/administración & dosificación , Factor VIIa/administración & dosificación , Hemorragia/tratamiento farmacológico , Animales , Tiempo de Sangría , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Estudios de Seguimiento , Hemodilución/efectos adversos , Hemodilución/métodos , Hemorragia/sangre , Hemorragia/inducido químicamente , Derivados de Hidroxietil Almidón/toxicidad , Sustitutos del Plasma/toxicidad , Conejos , Proteínas Recombinantes/administración & dosificación , Espectrofotometría , Tromboelastografía , Resultado del Tratamiento
19.
Br J Haematol ; 144(1): 99-106, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19016728

RESUMEN

The present study evaluated dynamic coagulation profiles, platelet aggregation, and thrombin generation in whole blood (WB) from eight children with thrombocytopenia during chemotherapy, and the haemostatic potential of platelets (+60 x 10(9)/l), recombinant factor VIIa (rFVIIa - NovoSeven), and a potent rFVIIa analogue (NN1731) both at 1 and 4 microg/ml. Dynamic WB coagulation profiles were recorded by thrombelastometry employing activation with tissue factor (TF - Innovin) at low concentrations. The baseline WB coagulation patterns were characterised by a prolonged clotting time (CT) and a pronounced reduction in clot propagation (MaxVel). WB platelet aggregation signal was five times lower in the study group compared with measurements in modelled thrombocytopenic WB from healthy volunteers. In vitro addition of fresh platelets reversed the coagulopathy. Addition of rFVIIa induced no significant changes in the thrombelastographic profile, whereas spiking with NN1731 shortened the CT significantly. The changes in WB thrombin generation reflected the changes in the MaxVel. In modeled thrombocytopenic WB from healthy individuals, both rFVIIa and NN1731 exhibited a pronounced haemostatic effect with NN1731 showing greater potency than rFVIIa. Compromised platelet function in the study group was assumingly responsible for the weakened haemostatic potential of rFVIIa as well as that of NN1731.


Asunto(s)
Factor VII/uso terapéutico , Factor VIIa/uso terapéutico , Hemostáticos/uso terapéutico , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/fisiopatología , Adolescente , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Agregación Plaquetaria , Proteínas Recombinantes/uso terapéutico , Tromboelastografía , Trombina/análisis , Trombocitopenia/sangre , Resultado del Tratamiento , Tiempo de Coagulación de la Sangre Total
20.
Thromb Haemost ; 101(5): 818-26, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19404533

RESUMEN

The mechanism for the elimination of factor VII (FVII) from the circulation is unknown, just as it is unclear how activation of FVII to FVIIa and subsequent complex formation with antithrombin III (AT) or alpha2-macroglobulin (alpha2M) affects clearance. The possibility that the clearance mechanism involves activation and inhibitor complex formation as obligatory intermediate reactions is examined in this study. Human and murine sera were spiked with human FVIIa in the absence and presence of heparin and analysed for complex formation. Complex formation in vivo was studied after intravenous injection of (125)I-VIIa in mice; and the pharmacokinetics (PK) of human and murine FVIIa was studied in normal mice. Furthermore, comparative PK studies were performed with FVII, FVIIa, active site blocked FVIIa and a preformed FVIIa-AT complex in normal and alpha2M-deficient mice. The data demonstrated that FVIIa-AT complexes and to a much lesser extent FVIIa-alpha2M-complexes accumulated in vivo after FVIIa administration. FVIIa-AT accounted for about 50% of total FVIIa antigen left in the circulation after 3 hours. All FVII derivatives studied including FVII, FVIIa and FVIIa-AT were cleared with similar rates suggesting an elimination kinetics which is unaffected by FVII activation and subsequent inactivation by plasma inhibitors.


Asunto(s)
Antitrombina III/metabolismo , Factor VII/farmacocinética , Factor VIIa/farmacocinética , Seroglobulinas/metabolismo , alfa-Macroglobulinas/metabolismo , Animales , Factor VIIa/administración & dosificación , Heparina/sangre , Humanos , Inyecciones Intravenosas , Radioisótopos de Yodo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Seroglobulinas/deficiencia , Seroglobulinas/genética , alfa-Macroglobulinas/deficiencia , alfa-Macroglobulinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA