The pharmacokinetic diversity of two von Willebrand factor (VWF)/ factor VIII (FVIII) concentrates in subjects with congenital von Willebrand disease

2011 ◽  
Vol 106 (08) ◽  
pp. 279-288 ◽  
Author(s):  
Craig M. Kessler ◽  
Friedman Ken ◽  
Bruce A. Schwartz ◽  
Joan C. Gill ◽  
Jerry S. Powell ◽  
...  

SummaryThe pharmacokinetic (PK) profiles of von Willebrand factor (VWF) /factor VIII (FVIII) concentrates are important for treatment efficacy and safety of von Willebrand disease (VWD) patients. This prospective, head-to-head, randomised crossover study compared the PK profile of a new, high purity, human plasma-derived (pd)VWF/FVIII concentrate, Wilate®, with the PK profile of an intermediate purity (pd)VWF/FVIII concentrate, Humate-P¯, in VWD patients. Subjects with inherited VWD were randomised to a single intravenous dose (40 IU/kg VWF ristocetin cofactor activity [VWF:RCo]) of Wilate® or Humate-P¯ in Period 1, and switched to the other study drug in Period 2. Each period was preceded by a washout time of ≥7 days. Coagulation factor parameters were analysed at multiple time-points. Of 22 randomised subjects, 20 had evaluable PK profiles, which indicated comparability for VWF antigen and VWF:RCo between Wilate® and Humate-P¯. The reported VWF:RCo average and terminal t1/2 of 10.4 and 15.8 hours (h), respectively, for Wilate® and 9.3 h and 12.8 h for Humate-P®, were not statistically different. Also, the mean VWF:RCo in vivo recoveries (Wilate® 1.89, Humate-P® 1.99 IU/dl per IU/kg) were similar between the two replacement therapies. Wilate® showed parallel decay curves for VWF:RCo and FVIII clotting activity (FVIII:C) over time, while FVIII:C of Humate-P® displayed a plateau between 0 and 12–24 h. This study demonstrated bioequivalent PK properties for VWF between Wilate® and Humate-P®. The PK profile of Wilate®, combined with the 1:1 VWF/FVIII ratio, theoretically should facilitate dosing and laboratory monitoring of VWF replacement to prevent bleeding in individuals with VWD.

Author(s):  
И.В. Куртов ◽  
Е.С. Фатенкова ◽  
Н.А. Юдина ◽  
А.М. Осадчук ◽  
И.Л. Давыдкин

Болезнь Виллебранда (БВ) может представлять определенные трудности у рожениц с данной патологией. Приведены 2 клинических примера использования у женщин с БВ фактора VIII свертывания крови с фактором Виллебранда, показана эффективность и безопасность их применения. У одной пациентки было также показано использование фактора свертывания крови VIII с фактором Виллебранда во время экстракорпорального оплодотворения. Von Willebrand disease presents a certain hemostatic problem among parturients. This article shows the effectiveness and safety of using coagulation factor VIII with von Willebrand factor for the prevention of bleeding in childbirth in 2 patients with type 3 von Willebrand disease. In one patient, the use of coagulation factor VIII with von Willebrand factor during in vitro fertilization was also shown.


Blood ◽  
2015 ◽  
Vol 125 (13) ◽  
pp. 2019-2028 ◽  
Author(s):  
Peter J. Lenting ◽  
Olivier D. Christophe ◽  
Cécile V. Denis

Abstract To understand the placement of a certain protein in a physiological system and the pathogenesis of related disorders, it is not only of interest to determine its function but also important to describe the sequential steps in its life cycle, from synthesis to secretion and ultimately its clearance. von Willebrand factor (VWF) is a particularly intriguing case in this regard because of its important auxiliary roles (both intra- and extracellular) that implicate a wide range of other proteins: its presence is required for the formation and regulated release of endothelial storage organelles, the Weibel-Palade bodies (WPBs), whereas VWF is also a key determinant in the clearance of coagulation factor VIII. Thus, understanding the molecular and cellular basis of the VWF life cycle will help us gain insight into the pathogenesis of von Willebrand disease, design alternative treatment options to prolong the factor VIII half-life, and delineate the role of VWF and coresidents of the WPBs in the prothrombotic and proinflammatory response of endothelial cells. In this review, an update on our current knowledge on VWF biosynthesis, secretion, and clearance is provided and we will discuss how they can be affected by the presence of protein defects.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 41-41 ◽  
Author(s):  
Patricia A. Lamont ◽  
Margaret V. Ragni

Abstract Although the extracellular association of Factor VIII (FVIII) and Von Willebrand Factor (VWF) is well established, the intracellular interaction of FVIII and VWF is not well understood. Recently, the importance of intracellular co-localization of FVIII and VWF for in vitro FVIII secretion was demonstrated in endothelial cell lines. Whether intracellular co-localization of FVIII and VWF is required for in vivo FVIII secretion, however, is not known. We previously showed that liver transplantation leads to phenotypic cure of hemophilia A, by virtue of FVIII production in the allograft liver. Because FVIII is synthesized only in the allograft liver but not in endothelial cells of transplant recipients, and VWF is synthesized in extrahepatic tissue, this is an ideal model to study whether co-localization of FVIII and VWF is required for in vivo FVIII secretion. We, therefore, studied FVIII and VWF response after desmopression (DDAVP) infusion, administered at 0.3 mcg/kg by intravenous infusion over 30 minutes, in each of two men with severe hemophilia A (FVIII:C <0.01 U/ml) who had undergone orthotopic liver transplantation for endstage liver disease six months earlier. Both men had HIV and hepatitis C co-infection and were clinically well, with mildly elevated liver function tests, and FVIII:C levels >30% following transplantation. Coagulation studies, drawn before and after DDAVP, revealed that VWF:RCoF and VWF:Ag, but not FVIII:C, increased after DDAVP administration (see Table). The prolonged aPTT and correction in a 1:1 aPTT mix confirmed the absence of an inhibitor in these subjects. The lack of FVIII response to DDAVP supports previous in vitro work, and demonstrates for the first time that intracellular co-localization of FVIII and VWF is essential for in vivo FVIII secretion. These data also suggest that extrahepatic FVIII synthesis is necessary for in vivo response of the DDAVP releasable pool of FVIII. By contrast, co-localization does not appear to be necessary for VWF secretion. Although it is not possible to exclude that a chronic, exhaustive post-transplant increase in VWF may have limited VWF response to DDAVP, it is clear that FVIII did not increase following DDAVP. These findings have important implications for the design of gene therapies for hemophilia A and Von Willebrand Disease. Subject Demographic Sample aPTT aPTT mix FVIII:C VWF:RCoF VWF:Ag 01-BW 32yoM Hem A Pre-DDAVP 44.4 sec 37.7 sec 0.50 U/ml 2.17 U/ml 2.42 U/ml HIV+/HCV+ Post-DDAVP 44.8 sec 37.4 sec 0.48 U/ml 2.91 U/ml 2.91 U/ml 02-PB 36yoM Hem A Pre-DDAVP 49.5 sec 38.0 sec 0.32 U/ml 1.61 U/ml 2.16 U/ml HIV+/HCV+ Post-DDAVP 50.8 sec 38.5 sec 0.30 U/ml 2.20 U/ml 2.50 U/ml


Blood ◽  
1997 ◽  
Vol 90 (9) ◽  
pp. 3555-3567 ◽  
Author(s):  
Peter L. Turecek ◽  
Herbert Gritsch ◽  
Ludwig Pichler ◽  
Wilfried Auer ◽  
Bernhard Fischer ◽  
...  

AbstractHereditary von Willebrand factor (vWF ) deficiency in Dutch Kooiker dogs, which have undetectable levels of vWF, causes spontaneous hemorrhage of mucosal surfaces similar to the clinical picture of von Willebrand disease in humans. Therefore, we used this canine model to study the in vivo effects of a new recombinant von Willebrand factor (rvWF ) preparation containing all species of vWF multimers compared with a rvWF fraction containing only low molecular weight multimers (LMW-rvWF ) and with a plasma-derived factor VIII/vWF concentrate (pdvWF ). In the vWF-deficient dogs, the half-life of vWF:Ag was 21.6 and 22.1 hours for rvWF, 7.7 hours for pdvWF, and 9 hours for LMW-rvWF; in vivo recovery of vWF:Ag was 59%, 64%, and 70% for rvWF, 33% for pdvWF and 92% for LMW-rvWF; in vivo recovery of RCoF was 78%, 110%, and 120% for rvWF, and 25% for pdvWF. Both rvWF and pdvWF caused increases in factor VIII, which were sustained even when vWF:Ag had decreased to nearly undetectable levels and only monomeric or dimeric species were detectable on agarose gels. At the dosages used, no effect was seen on bleeding time, but the rate of blood flow from cuticle wounds was reduced after a single bolus administration of rvWF. The rvWF was able to control a severe nose bleed in one dog.


Blood ◽  
2014 ◽  
Vol 124 (3) ◽  
pp. 445-452 ◽  
Author(s):  
Andrew Yee ◽  
Robert D. Gildersleeve ◽  
Shufang Gu ◽  
Colin A. Kretz ◽  
Beth M. McGee ◽  
...  

Key Points The D′D3 domains of VWF are sufficient to stabilize FVIII in vivo. The prolongation of VWF D′D3 survival in vivo by Fc fusion elevates FVIII levels in the setting of VWF but not FVIII deficiency.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3672-3672 ◽  
Author(s):  
Craig M. Kessler ◽  
Jerry Powell ◽  
Bruce A. Schwartz

Abstract Abstract 3672 Objective: Understanding pharmacokinetic (PK) profiles of von Willebrand Factor (VWF)/Factor VIII (FVIII) concentrates is important in the treatment of VWD. This prospective, randomized, open-label, crossover trial investigated the PK characteristics of two plasma-derived (pd) VWF/FVIII concentrates (Wilate® [high-purity (HP)] and Humate-P® [intermediate-purity (IP)]) in subjects with inherited VWD. Methods: After a wash-out period, twenty-two subjects (Type 1, n=6; Type 2, n=9 [6 Type 2A, 1 Type 2B, and 2 Type 2M]; and Type 3, n=7) were randomized in a prospective, controlled, open-labeled, 2-arm crossover, to receive 40 IU VWF:RCo/kg of Wilate® or Humate-P® followed by another washout period and administration of the other study drug. Blood samples were taken at multiple time-points over 72hrs for PK evaluations. Results: A non compartmental statistical model was used to analyze the data. The mean VWF:RCo half-life for Type-3 subjects was similar for the HP product (9.1 hours [±2.6]) and IP product (10.2 hours [±2.1]. For all study participants there were no significant differences in incremental in-vivo recoveries (≂f2.0 IU VWF:RCo/dL per IU/kg), or in clearance. Bioequivalence between the two products was shown for the main VWF PK parameters. The decay-curves for VWF:RCo and FVIII:C in the HP product showed a parallel decay, while the IP product did not and showed an unusually sustained FVIII plateau over the initial decay time period. The data showed an almost a 2 fold decrease in clearance between the two products. The chromogenic FVIII:C terminal half live for the two products were similar (16.1hrs [SD=3.1] HP product and 20.5 hrs [SD=7.6] IP product), although the average half lives were quite different (13 hrs [SD=4.1] HP product and 37.7 hrs [SD=6.4] IP product). This was in spite of the much higher initial FVIII concentration profile of the 1:1 VWF/FVIII HP product. The 2.4:1 VWF/FVIII IP product showed an expected lower mean FVIII:C peak value due to the excess of VWF:RCo activity in the product that was based on VWF:RCo unit dosing, but similar both products had a similar dose adjusted IVR. Conclusions: This study confirms the similarity of the VWF PK properties of the two licensed VWF products. However, the analysis of FVIII activity, in the two products revealed some differences in their PK profiles. With the similar PK-properties for VWF:RCo and FVIII:C, Wilate showed a more parallel course for the two active components. A more predictable PK profile for both VWF and FVIII in a product may facilitate both more accurate dosing and laboratory monitoring of VWF replacement in VWD treatment. A parallel PK profile together with an equal ratio for FVIII and VWF activities may help avoid over or under dosing of either of the two critical coagulation parameters. Disclosures: Kessler: Grifols S.A.: Research Funding. Schwartz:Octapharma: Employment.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 1-1
Author(s):  
Amber Federizo

Inherited platelet disorders are recognized as an important cause of mild to severe bleeding in both children and adults. Patients with platelet disorders may present with mucocutaneous bleeding, gastrointestinal bleeding, menorrhagia, postsurgical, and/or excessive bleeding from traumatic injury. Delta storage pool deficiencies (delta-SPD) are among the most frequent platelet disorders, characterized by dysfunctional dense platelet granules. Bernard Soulier syndrome (BSS) is an autosomal recessive platelet disorder caused by mutations in various polypeptides in the GpIb/IX/V complex, which is the principal receptor for von Willebrand factor (VWF). Treatment of platelet disorders is mainly supportive. Normal hemostasis requires VWF and factor VIII (FVIII) to support platelet adhesion and aggregation at sites of vascular injury. von Willebrand factor is a large multimeric glycoprotein present in human plasma as a series of polymers called multimers. Molecular weights for multimers ranges from 500 kDa for the dimer to over 10,000 kDa for the high molecular weight multimers (HMWM) forming the largest known protein present in human plasma. Each multimeric subunit of VWF has binding sites for the receptor GpIb on nonactivated platelets and the receptor GpIIb/IIIa to facilitate platelet adhesion and platelet aggregation, respectively, making the VWF HMWM important for normal platelet function. Desmopressin (DDAVP), which is known to stimulate the release of VWF and FVIII, is commonly used for treatment of platelet disorders. Potentiation of platelet aggregation at high shear rate may be one mechanism by which DDAVP shortens the prolonged bleeding time of patients with congenital platelet defects. For severe bleeding, platelet transfusion may be required, but patients may develop isoantibodies, rendering this therapy ineffective. For this reason, it may be prudent to reserve platelet transfusion in this patient population for emergent situations, such as trauma. Other patients and/or clinical situations may require recombinant active factor VII (rFVIIa), but this therapy is very costly and not always effective and/or available. Antifibrinolytics may also be used but are not always effective. In four (4) patients with platelet disorders (delta-SPD [n=3]; BSS [n=1]), common supportive therapies were not effective, tolerable, and/or available. It was postulated that off-label infusions of a cost-effective von Willebrand factor/coagulation factor VIII (VWF/FVIII) complex (Wilate, Octapharma SA) might be of benefit in these refractory patients (Table 1). The mechanism of action of DDAVP treatment efficacy relies on the release of existing, stored, functional VWF. In refractory patients with suboptimal VWF functionality, it was reasoned that infusion of exogenous, functional VWF and FVIII could potentially encourage platelet adhesion and aggregation. All refractory patients studied were treated successfully with the VWF/FVIII complex with positive clinical outcomes. As mentioned, the adhesive activity of VWF depends on the size of its multimers, and HMWM are the most effective in supporting interaction with collagen and platelet receptors and in facilitating wound healing under conditions of shear stress in the human vascular system. The VWF/FVIII complex utilized in these patients is known to have minimal amounts of the plasma metalloproteinase ADAMTS13. The HMWM of VWF are, under normal conditions, cleaved by ADAMTS13 to smaller, less adhesive multimers. During the manufacturing process, if the ADAMTS13 is not filtered out of the product almost entirely, the VWF in the vial may become highly proteolyzed. Therefore, a reduction or lack of HMWM resulting from inclusion of ADAMTS13 in the manufactured product is believed to reduce product functionality. Multimeric analysis of the VWF/FVIII complex has shown that it exhibits a physiological triplet structure which resembles normal plasma. In addition, the product has a high safety profile and tolerability as protein impurities are eliminated in the manufacturing process. In summary, the use of a VWF/FVIII complex in four (4) patients with inherited platelet disorders, who were refractory to conventional treatments, provided beneficial, cost-effective clinical outcomes with resolution of bleeding. Disclosures Federizo: Octapharma: Consultancy, Honoraria, Other: Publication support, Speakers Bureau; Sanofi: Consultancy, Honoraria, Research Funding, Speakers Bureau; American Thrombosis and Hemostasis Netowrk: Research Funding; Aptevo: Consultancy, Speakers Bureau; National Hemophilia Foundation: Consultancy, Honoraria. OffLabel Disclosure: von Willebrand/FVIII concentrate is currently approved for the treatment of Hemophilia A and von Willebrand. This abstract will review the off-label use of this medication in the treatment of inherited platelet dysfunction.


F1000Research ◽  
2018 ◽  
Vol 7 ◽  
pp. 96 ◽  
Author(s):  
Karl C Desch

Von Willebrand factor (VWF) is a multimeric plasma glycoprotein that plays a central role in the initiation of blood coagulation. Through interactions between its specific functional domains, the vascular wall, coagulation factor VIII, and platelet receptors, VWF maintains hemostasis by binding to platelets and delivering factor VIII to the sites of vascular injury. In the healthy human population, plasma VWF levels vary widely. The important role of VWF is illustrated by individuals at the extremes of the normal distribution of plasma VWF concentrations where individuals with low VWF levels are more likely to present with mucocutaneous bleeding. Conversely, people with high VWF levels are at higher risk for venous thromboembolic disease, stroke, and coronary artery disease. This report will summarize recent advances in our understanding of environmental influences and the genetic control of VWF plasma variation in healthy and symptomatic populations and will also highlight the unanswered questions that are currently driving this field of study.


Sign in / Sign up

Export Citation Format

Share Document