Development and Characterization of Recombinant Ovine Factor VIII

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1193-1193
Author(s):  
Philip M Zakas ◽  
Bagirath Gangadharan ◽  
Graca Almeida-Porada ◽  
Christopher D Porada ◽  
H. Trent Spencer ◽  
...  

Abstract Abstract 1193 Factor VIII (fVIII) is an essential glycoprotein procofactor in the blood coagulation cascade. Mutations in the fVIII gene often result in diminished plasma fVIII activity causing the bleeding disorder hemophilia A, which obeys X-linked recessive genetics and affects approximately 1 in 7500 males. Current treatment is limited to intravenous infusion of plasma-derived or recombinant human (h)-fVIII containing products. This therapy is expensive and requires multi-weekly injections to achieve prophylaxis, which must be maintained for the duration of the patients' life to avoid debilitating joint disease as well as life-threatening bleeding episodes. While gene therapy is being explored as a potential cure, much research is aimed at improving the therapeutic utility of recombinant fVIII. Our laboratory has been interested in the comparison of recombinant fVIII molecules derived from different animal species and has identified many species-specific biochemical properties. The characterization of recombinant murine (m)-factor VIII revealed near complete stability at physiologic concentrations following thrombin activation upon which h-fVIII activity decays on the order of minutes. In contrast, porcine (p)-fVIII demonstrates 100-fold increased post-translational biosynthesis over h-fVIII as well as decreased engagement of the endoplasmic reticulum-resident unfolded protein response. Arruda and colleagues showed that canine (c)-fVIII displays 3-fold higher specific activity than that of h-, p-, or m-fVIII and currently is being used to treat bleeding episodes in canine hemophilia A colonies. Finally, we have generated hybrid fVIII constructs, mapped the sequences necessary and sufficient for certain differential properties, identified immunogenic epitopes inherent to each species and created novel fVIII molecules with combined potentially beneficial characteristics. The current study is a continuation of this line of research focusing on ovine (o)-fVIII. Recently, a line of hemophilia A sheep was re-established and the pathology and clinical profile was described. O-fVIII possesses a high degree of homology to h-fVIII with a similar A1-A2-activation peptide-A3-C1-C2 domain structure. The causative mutation was identified as a single nucleotide insertion causing a frameshift and premature stop codon in exon 14. Administration of human and hybrid h/p-fVIII in these sheep corrected the bleeding phenotype transiently, but invariably induced the formation of high titer anti-fVIII inhibitory antibodies leading to premature mortality. Herein, we describe the generation, expression and biochemical characterization of recombinant full-length and B-domain-deleted (BDD) o-fVIII. O-fVIII was cloned into the mammalian expression vector, ReNeo and a pace/furin linker sequence was introduced between the A2 domain and the activation peptide. Utilizing a baby hamster kidney cell expression system, full-length o- and h-fVIII demonstrated similar expression levels at 0.098 ± 0.024 and 0.022 ± 0.006 units/106 cells/24hr, respectively (p = 0.99). Removal of the ovine B domain resulted in increased expression (0.97 ± 0.2 units/106 cells/24hr, p < 0.001) to a level equivalent to BDD h-fVIII (0.49 ± 0.149 units/106 cells/24hr; p = 0.06). BDD o-fVIII was purified to virtual homogeneity from conditioned serum-free media using a two-step ion-exchange chromatography procedure identified previously for the purification of recombinant h- and p-fVIII. Two independent preparations were analyzed and determined to have specific activities of 12,300 and 14,760 units/mg. SDS-PAGE analysis revealed three predominant polypeptides species, with a minority of intact single chain as well as predominantly processed heavy and light chains. Uniquely, the heavy and light chain polypeptides displayed similar relative mobility and could only be distinguished by thrombin proteolysis or immuno-precipitation prior to SDS-PAGE using monoclonal antibodies with known epitopes in either the heavy or light chains. We anticipate that recombinant o-fVIII, similar to the situation that has occurred with the canine hemophilia A colony and recombinant c-fVIII, will facilitate the maintenance of the ovine hemophilia A herd and their utilization as a relevant large animal model for research and development of novel hemophilia A therapeutics. Disclosures: No relevant conflicts of interest to declare.

1997 ◽  
Vol 78 (05) ◽  
pp. 1352-1356 ◽  
Author(s):  
Emel Aygören-Pürsün ◽  
Inge Scharrer ◽  

SummaryIn this open multicenter study the safety and efficacy of recombinant factor VIII (rFVIII) was assessed in 39 previously treated patients with hemophilia A (factor VIII basal activity ≤15%).Recombinant FVIII was administered for prophylaxis and treatment of bleeding episodes and for surgical procedures. A total of 3679 infusions of rFVIII were given. Efficacy of rFVIII as assessed by subjective evaluation of response to infusion and mean annual consumption of rFVIII was comparable to that of plasma derived FVIII concentrates. The incremental recovery of FVIII (2.4 ± 0,83%/IU/kg, 2.12 ± 0.61%/IU/kg, resp.) was within the expected range. No clinical significant FVIII inhibitor was detected in this trial. Five of 16 susceptible patients showed a seroconversion for parvovirus B19. However, the results are ambiguous in two cases and might be explained otherwise in one further case. Thus, in two patients a reliable seroconversion for parvovirus B19 was observed.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5050-5050
Author(s):  
Mark J. Belletrutti ◽  
Roxanne Seiferman-Nelson ◽  
Bonny Granfield

Abstract Introduction: Development of circulating anti-factor VIII antibodies (inhbitors) is the most serious and challenging complication in the treatment of hemophilia A. Up to 38% of hemophilia patients develop inhibitors with recombinant FVIII (rFVIII) products (Gouw et al. N Engl J Med. 2013; 368:231-239). The presence of inhibitors leads to an increased risk of bleeding, poor physical functioning and quality of life (Benson et al., Eur. J. Haematol. 2012; 88:371-379). Immune tolerance induction (ITI) is the most common method for eliminating inhibitors, historically performed with high dose, and prolonged treatment with plasma-derived (pd), or recombinant FVIII (rFVIII) concentrates. Although ITI for the eradication of inhibitors has become standard of care for hemophilia patients the therapeutic superiority of a particular product type (rFVIII vs. pd-FVIII) has not yet been conclusively demonstrated. In accordance with its role in stabilizing FVIII, the presence of von Willebrand factor (VWF) in pd-FVIII concentrates has been shown to improve the outcome of ITI. Wilate® (Octapharma) is a high-purity human plasma derived complex containing two proteins (VWF and FVIII) in a 1:1 ratio. The aim of this study was to determine the effectiveness of Wilate for primary ITI therapy for six patients with severe hemophilia A. Patients and Methods: The case history for six pediatric hemophilia A patients prior to and during primary Wilate ITI was reviewed. For 5/6 patients, inhibitors developed during rFVIII factor replacement therapy. For the sixth patient, inhibitors were detected at the time of hemophilia diagnosis. ITI began once patients achieved an inhibitor titer of less than 10 BU/mL. The ITI dosing regimen ranged from 50-60 IU/Kg of Wilate three times per week to 200 IU/Kg once daily. Inhibitor titers were measured regularly, prior to and during ITI using the Nijmegen-Bethesda assay. The number of port-a-cath infections and bleeding episodes were also monitored. ITI success was defined as: an undetectable inhibitor level (<0.6 BU/mL), FVIII plasma recovery ≥ 66% of predicted, and FVIII half-life ≥6 hours. Results: Wilate ITI was well tolerated in all patients, with no product-related adverse events. All patients had a port-a-cath device inserted for Wilate injections. Two port-a-cath infections occurred during ITI. Five of six patients had poor prognostic factors for ITI outcome. These poor prognostic factors included a high-risk FVIII gene mutation, historical peak inhibitor titer greater than 50 BU/mL, age of ITI onset greater than 6 years, and ITI onset more than 12 months from inhibitor development. The frequency of these poor prognostic factors varied amongst the patients: 1 patient had 4, 1 patient had 2, and 3 patients presented with 1 poor prognostic factor. Despite the presence of these high-risk factors, Wilate was successful at reducing the inhibitor titers to undetectable levels in all patients. Furthermore, inhibitor titers have remained low or undetectable without significant spikes for the duration of treatment. Patient plasma recovery and FVIII half-life results have also indicated that patients are progressing towards successful ITI. Importantly, for 6/6 patients (including 3 patients who had previously been treated with Anti-Inhibitor Coagulant Complex (FEIBA) prophylaxis therapy) - Wilate therapy was successful at reducing the number of bleeding episodes allowing for the cessation of FEIBA prophylaxis. Since commencing Wilate ITI, 6/6 patients have not reported any major bleeding episodes. The improved clinical outcome was perceived by the patients as an improved well-being, and quality of life. Conclusion: Wilate ITI was found to be well tolerated, safe, and successful at reducing inhibitor levels to below the detectable range for six severe hemophilia A patients. Patients experienced no treatment related adverse events, had a low rate of port-a-cath infections, and did not present with any major bleeding episodes while on Wilate ITI. In light of the 3-5 fold increase in overall treatment costs of immune tolerance induction, careful consideration should be given to choice of product (rFVIII versus pd-FVIII) – especially for patients at high-risk of failure. (Dimichele et al. Haemophilia 2004: 10 Suppl 4;140-145). The present data suggest that Wilate, a pd-FVIII product, is effective in managing patients with inhibitors. Disclosures Belletrutti: Baxter Canada: Honoraria, Membership on an entity's Board of Directors or advisory committees; CSL Behring Canada: Honoraria.


Blood ◽  
2006 ◽  
Vol 108 (12) ◽  
pp. 3668-3673 ◽  
Author(s):  
Jack Spira ◽  
Olga P. Plyushch ◽  
Tatyana A. Andreeva ◽  
Yury Andreev

AbstractProphylactic treatment for hemophilia A involves infusion of factor VIII (FVIII) concentrates every 2 to 3 days. Liposomes can be efficacious vehicles for medicines, and surface modification by PEGylation can prolong liposome circulation time. When reconstituted with PEGylated liposomes (PEGLip's), recombinant FVIII binds noncovalently but with high affinity to the external liposome surface. This preparation showed prolongation of FVIII half-life and increased protection from bleeding in preclinical models. Here we report a blinded, controlled, crossover, multicenter clinical study that evaluated the time free from bleeding episodes in patients with hemophilia A during prophylaxis with standard rFVIII (no liposomes) or PEGLip rFVIII (PEGLip reconstituted) at 25 and 35 IU/kg doses. Of 24 enrolled patients, 23 were eligible for efficacy analysis. Mean number of days without bleeds was 7.2 ± 1.7 with standard rFVIII compared with 13.3 ± 4.8 with PEGLip rFVIII at the 35 IU/kg dose and 5.9 ± 1.7 with standard rFVIII versus 10.9 ± 2.9 with PEGLip rFVIII at the 25 IU/kg dose (P < .05 between treatment groups for each dose). PEGLip rFVIII was well tolerated. These data suggest that reconstitution of rFVIII with PEGLip's may reduce the frequency of treatment during prophylaxis.


Blood ◽  
1976 ◽  
Vol 48 (6) ◽  
pp. 931-939
Author(s):  
JM Lavergne ◽  
D Meyer ◽  
H Reisner

Anti-factor VIII antibodies from three hemophilia A patients were selectively isolated by immune complex formation and secondary precipitation with polyethylene glycol. These antibodies were characterized as IgG, with only one type of light chains (kappa) and a mixture of IgG3 and IgG4 heavy chains. This finding indicated that stable complexes of factor VIII and homologous anti-factor VIII antibodies could be obtained, and that the immunologic response to the infusion of homologous factor VIII was oligoclonal rather than monoclonal in these patients.


2021 ◽  
Vol 47 (1) ◽  
Author(s):  
Baoyu Lei ◽  
Chuang Liang ◽  
Haiyan Feng

Abstract Background Congenital hemophilia A is a recessive inherited hemorrhagic disorder. According to the activity of functional coagulation factors, the severity of hemophilia A is divided into three levels: mild, moderate and severe. The first bleeding episode in severe and moderate congenital hemophilia A occurs mostly in early childhood and mainly involves soft tissue and joint bleeds. At present, there are limited reports on severe congenital hemophilia A with low factor XII (FXII) activity during the neonatal period. Case presentation A 13-day-old neonate was admitted to the hospital with hematoma near the joints of both upper arms. Coagulation tests showed he had low activity of factor VIII (FVIII) and FXII. He was diagnosed with congenital hemophilia A and treated with human coagulation factor VIII (recombinant FVIII). Although the hematoma became smaller, FVIII activity was only increased to a certain extent and FXII activity decreased gradually. Unfortunately, the child responded poorly to recombinant human coagulation factor VIII and his guardian rejected prophylactic inhibitors and genetic testing and refused further treatment. Three months later, the child developed intracranial hemorrhage (ICH) due to low FVIII activity. Conclusions In hemophilia A, the presence of FVIII inhibitors, drug concentration and testing are three important aspects that must be considered when FVIII activity does not reach the desired level. Early positive disease treatment and prophylaxis can decrease the frequency of bleeding and improve quality of life. We recommend that pregnant women with a family history of hemophilia A undergo early prenatal and neonatal genetic testing.


Blood ◽  
2020 ◽  
Vol 136 (22) ◽  
pp. 2524-2534 ◽  
Author(s):  
Steffen Rosen ◽  
Stefan Tiefenbacher ◽  
Mary Robinson ◽  
Mei Huang ◽  
Jaydeep Srimani ◽  
...  

Abstract Adeno-associated virus (AAV)-based gene therapies can restore endogenous factor VIII (FVIII) expression in hemophilia A (HA). AAV vectors typically use a B-domain–deleted FVIII transgene, such as human FVIII-SQ in valoctocogene roxaparvovec (AAV5-FVIII-SQ). Surprisingly, the activity of transgene-produced FVIII-SQ was between 1.3 and 2.0 times higher in one-stage clot (OS) assays than in chromogenic-substrate (CS) assays, whereas recombinant FVIII-SQ products had lower OS than CS activity. Transgene-produced and recombinant FVIII-SQ showed comparable specific activity (international units per milligram) in the CS assay, demonstrating that the diverging activities arise in the OS assay. Higher OS activity for transgene-produced FVIII-SQ was observed across various assay kits and clinical laboratories, suggesting that intrinsic molecular features are potential root causes. Further experiments in 2 participants showed that transgene-produced FVIII-SQ accelerated early factor Xa and thrombin formation, which may explain the higher OS activity based on a kinetic bias between OS and CS assay readout times. Despite the faster onset of coagulation, global thrombin levels were unaffected. A correlation with joint bleeds suggested that both OS and CS assay remained clinically meaningful to distinguish hemophilic from nonhemophilic FVIII activity levels. During clinical development, the CS activity was chosen as a surrogate end point to conservatively assess hemostatic efficacy and enable comparison with recombinant FVIII-SQ products. Relevant trials are registered on clinicaltrials.gov as #NCT02576795 and #NCT03370913 and, respectively, on EudraCT (European Union Drug Regulating Authorities Clinical Trials Database; https://eudract.ema.europa.eu) as #2014-003880-38 and #2017-003215-19.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1078-1078
Author(s):  
Sabine Pestel ◽  
Philipp Claar ◽  
Jochen Müller-Cohrs ◽  
Elmar Raquet ◽  
Hendrik Peil ◽  
...  

Abstract Introduction: Current state of the art treatment of severe Hemophilia A is prophylactic or on-demand replacement of the deficient factor VIII (FVIII) using plasma derived or recombinant factors [Keeling 2008; Srivastava 2012; Wong 2011]. The compliance to existing therapies is hampered by the short half-life of available FVIII products necessitating multiple intravenous injections per week for adequate prophylaxis [Bjorkman 2009; Sheridan 2011]. Thus, FVIII products with improved pharmacokinetic profiles are currently under development or have recently been made available [Oldenburg 2014]. FVIII's pharmacokinetic (PK) properties are believed to be strongly dictated by its binding to von Willebrand Factor (VWF) [Lenting 2007; Dasgupta 2007; Saenko 2006]. The aim of this study was to compare the binding affinity of different recombinant FVIII (rFVIII) products to plasma derived VWF (pd VWF), and characterize their PK properties in different animal species. Methods: The PK profiles of rVIII-SingleChain (B-domain-truncated, covalently-linked single chain rFVIII; CSL Behring), Advate® (full-length rFVIII; Baxalta), NovoEight® (B-domain-truncated rFVIII; NovoNordisk), Eloctate® (B-domain-deleted rFVIII fused to Fc portion of IgG; Biogen) and Nuwiq® (B-domain-deleted rFVIII; Octapharma) were compared in FVIII knock out mice (FVIII ko), rats and rabbits following intravenous injection of 150 (mice and rabbits) or 400 or 480 (rats) IU/kg rFVIII. Dose levels were administered based on labelled/nominal chromogenic FVIII activity. Plasma samples were obtained up to 72 h (mice), 48 h (rats) and 96 h (rabbits) post-application. FVIII plasma levels were determined based on antigen (FVIII:Ag) using an ELISA technique (ASSERACHROM®, Stago) and/or based on chromogenic FVIII activity (FVIII:C; COAMATIC® FACTOR VIII, Chromogenix). Results: The PK of rFVIII products in three different animal species was compared using the area under the plasma concentration time curve (AUC) as an indicator for exposure. In FVIII ko mice, FVIII:Ag based PK of rVIII-SingleChain was comparable to that of Eloctate® and NovoEight®, and superior over Nuwiq® and Advate® based on AUC. In contrast, FVIII:C based PK data did not achieve significant differences, most likely due to the the higher assay variability. In rats, FVIII:Ag PK of rVIII-SingleChain was superior over all tested rFVIII products, i.e. Nuwiq®, NovoEight®, Eloctate® and Advate® based on AUC. In rabbits, rVIII-SingleChain FVIII:Ag PK was comparable to NovoEight® and superior to all other rFVIII products, i.e. Nuwiq®, Eloctate® and Advate®. Thus, across all animal species tested, a favorable PK profile was shown for rVIII-SingleChain. It was further shown that the binding affinity of rVIII-SingleChain to pd VWF was significantly improved in comparison to the full length molecule Advate® [Zollner 2014]. This higher binding affinity to pd VWF may therefore explain the differences in PK profiles observed. Conclusion: In general, rVIII-SingleChain showed a favorable PK profile as compared to other rFVIII products such as Advate®, NovoEight®, Eloctate® and Nuwiq® across species, i.e. FVIII ko mice, rats and rabbits. This favorable PK profile of rVIII-SingleChain may be explained by its high binding affinity to VWF. Disclosures Pestel: CSL Behring GmbH: Employment. Claar:CSL Behring GmbH: Employment. Müller-Cohrs:CSL Behring GmbH: Employment. Raquet:CSL Behring GmbH: Employment. Peil:CSL Behring GmbH: Employment. Weimer:CSL Behring GmbH: Employment. Herzog:CSL Behring GmbH: Employment.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1025-1025
Author(s):  
Jolanta Krudysz-Amblo ◽  
Behnaz Parhami-Seren ◽  
Saulius Butenas ◽  
Kathleen E. Brummel-Ziedins ◽  
Edward D. Gomperts ◽  
...  

Abstract Hemophilia A is a clinically heterogeneous bleeding disorder characterized by the absence of functional factor VIII (FVIII). The most severe complication from exposure to exogenous factor VIII therapy is the development of alloantibodies that neutralize the effect of the therapeutic agent. The standard method for quantitation of FVIII inhibitors in human plasma is by the Nijmegen method, a modification of the Bethesda assay. Our objective was to develop a sensitive and specific fluorescence based immunoassay for the quantitation of anti-FVIII antibodies (FVIIIAb) in human plasma. An affinity purified human anti-FVIIIAb isolated from a hemophilia A patient was used as a calibrator, with a detectability limit of 40±1.5pM. The calibrator and the human plasma anti-FVIIIAb were captured on recombinant FVIII coupled microspheres and probed with mouse anti-human Ig conjugated to R-Phycoerythrin. Citrated human plasma samples from 150 healthy donors and 39 inhibitor-negative hemophilia A subjects were studied and compared to 4 inhibitor-positive hemophilia A plasma samples with inhibitor titers of 1BU (94.6±0.08nM), 11BU (214.3±7.1nM), 106BU (2209.4±84.9nM) and 140BU (2417.7±3.8nM) as measured by the Nijmegen method. Among the 150 normal control plasmas, 4 subjects (3%) were determined as positive (anti-FVIIIAb ≥ 40pM) with concentrations in the range of 0.6nM to 1.9nM, well below that of 1BU (94.6±0.08nM) inhibitor-positive sample. Anti-FVIIIAb was also evaluated in seven commercial inhibitor-free plasma samples reported to contain &lt;1% FVIII activity. Two of the seven plasmas were found to contain anti-FVIIIAb with concentrations of 112.9±1.9nM and 71.1±8.5nM. The concentration of the antibody in both samples was within the range of the measured molar concentration of an inhibitor-positive 1BU titer plasma sample. Thirty two inhibitor-negative hemophilia A subjects were also quantitated for anti-FVIIIAb. Eleven of the 32 samples tested positive for anti-FVIIIAb with molar concentrations in the range of 0.6–20nM. The overall prevalence of the non-inhibitory anti-FVIIIAbs in hemophilia A subjects evaluated by our immunoassay (n=39) is approximately 33% relative to healthy donor population in which 3% were determined as positive (≥40pM) for FVIIIAb.


Blood ◽  
1976 ◽  
Vol 48 (6) ◽  
pp. 931-939 ◽  
Author(s):  
JM Lavergne ◽  
D Meyer ◽  
H Reisner

Abstract Anti-factor VIII antibodies from three hemophilia A patients were selectively isolated by immune complex formation and secondary precipitation with polyethylene glycol. These antibodies were characterized as IgG, with only one type of light chains (kappa) and a mixture of IgG3 and IgG4 heavy chains. This finding indicated that stable complexes of factor VIII and homologous anti-factor VIII antibodies could be obtained, and that the immunologic response to the infusion of homologous factor VIII was oligoclonal rather than monoclonal in these patients.


Sign in / Sign up

Export Citation Format

Share Document