von Willebrand factor (VWF) is a large multimeric adhesive glycoprotein with complex roles in thrombosis and hemostasis. Abnormalities in VWF give rise to a variety of bleeding complications, known as von Willebrand disease (VWD), the most common inherited bleeding disorder in humans. Current treatment of VWD is based on the replacement of the deficient or dysfunctional protein either by endogenous release from endothelial Weibel-Palade bodies or by administration of plasma-derived VWF concentrates. During the last years, several efforts have been made to optimize existing therapies for VWD, but also to devise new approaches, such as inducing endogenous expression with interleukin-11, administering exogenous recombinant VWF, or introducing the protein via gene delivery. Clearly, the efficacy of any strategy will depend on several factors, including, for example, the quantity, activity, and stability of the delivered VWF. The inherent complexity of VWF biosynthesis, which involves extensive posttranslational processing, may be limiting in terms of producing active VWF outside of its native cellular sources. This review summarizes recent progress in the development of different treatment strategies for VWD, including those that are established and those that are at the experimental stage. Potential pitfalls and benefits of each strategy are discussed.

In 1926, Finnish physician Erik von Willebrand described an inherited bleeding disorder that was distinct from hemophilia A.1  It took another 30 years before the plasma protein that is central to the disease was identified, and this was eponymously named von Willebrand factor (VWF).2,3  Since then, much of the structure and function of VWF have been elucidated, and insights into the pathology of von Willebrand disease (VWD) have been continuously gained. This has naturally led to the development of improved treatment strategies for this sometimes life-threatening disorder. In this review, we discuss the different treatment modalities of VWD, focusing on both clinically established regimens and experimental strategies.

Although not recognized at that time, the bleeding problems originally described by von Willebrand illustrate the pivotal role of VWF in hemostasis. Indeed, this large, multimeric plasma protein is essential for the recruitment of circulating platelets at sites of vascular injury under high shear conditions. Furthermore, apart from sustaining platelet adhesion, VWF fulfills a crucial role in hemostasis by protecting coagulation factor VIII (FVIII) from rapid degradation, cellular uptake, or binding to the surface of activated platelets and endothelial cells. Indeed, when not bound to VWF, the plasma half-life of FVIII is reduced from 12 hours to 1 to 2 hours.

The multidomain structure of VWF (D1-D2-D′-D3-A1-A2-A3-D4-B1-B2-B3-C1-C2-CK)4,5  is fundamental to its function (Figure 1).6,7  At sites of vascular injury, VWF is able to bind to exposed fibrillar collagens via its A3 domain.8,9  This, and/or the high shear stress exerted on immobilized VWF, induces exposure of the VWF A1 domain because of loss of the shielding effect of the A2 and/or D′D3 domain.10-12  These conformational changes allow adhesion of platelets from the circulation via interactions with platelet glycoprotein (GP)Ibα. Because of the fast on- and off-rates of the GPIbα-VWF interaction,13-17  platelets are decelerated, allowing the establishment of more firm interactions between the platelet collagen receptors and the exposed subendothelium. Platelet adhesion is then followed by platelet aggregation, which involves interactions of activated platelet integrin αIIbβ3 with the RGD sequence found in fibrinogen and in the C1 domain of VWF.

Figure 1

Schematic representation of VWF. VWF is synthesized as a pre-pro-VWF that comprises a 22-residue signal peptide, a 741-residue propeptide, and the 2050-residue mature subunit. After removal of the signal peptide (SP), pro-VWF subunits associate in the endoplasmic reticulum in “tail-to-tail” dimers by the formation of disulfide bonds between the cysteine-rich carboxyl-terminal CK domains, after which dimers further multimerize by forming “head-to-head” disulfide bonds between the amino-terminal cysteine-rich D3 domains in the Golgi. The propeptide and the mature subunit form pro-VWF (2791 residues) consisting of 4 types of repeated domains as indicated. Crystal structures for the A16  and A37  domains are depicted. The main binding sites that are important for the hemostatic function of VWF are indicated together with the ADAMTS13 cleavage site. Major regions in which mutations have been found that are associated with VWD types 1 and 2 are also shown.

Figure 1

Schematic representation of VWF. VWF is synthesized as a pre-pro-VWF that comprises a 22-residue signal peptide, a 741-residue propeptide, and the 2050-residue mature subunit. After removal of the signal peptide (SP), pro-VWF subunits associate in the endoplasmic reticulum in “tail-to-tail” dimers by the formation of disulfide bonds between the cysteine-rich carboxyl-terminal CK domains, after which dimers further multimerize by forming “head-to-head” disulfide bonds between the amino-terminal cysteine-rich D3 domains in the Golgi. The propeptide and the mature subunit form pro-VWF (2791 residues) consisting of 4 types of repeated domains as indicated. Crystal structures for the A16  and A37  domains are depicted. The main binding sites that are important for the hemostatic function of VWF are indicated together with the ADAMTS13 cleavage site. Major regions in which mutations have been found that are associated with VWD types 1 and 2 are also shown.

Close modal

The biosynthesis of VWF is restricted to endothelial cells and megakaryocytes and is complex, involving removal of the signal peptide and the propeptide, glycosylation, sulphatation, and multimerization. Endothelial VWF is secreted constitutively in the blood or in the subendothelial matrix. It may also be stored in Weibel-Palade bodies, from which it is released either on stimulation or via an unstimulated basal pathway.18  After biosynthesis in megakaryocytes, VWF accumulates in platelet α-granules and is only released via a regulated pathway. No matter what the cellular source, VWF activity is dependent on the extent and pattern of multimerization. Local secretion of ultra-large (UL, > 10 × 106 Da) VWF multimers from Weibel-Palade bodies and α-granules at sites of vascular injury strategically localizes highly active VWF to effect hemostasis. Nevertheless, because these UL-VWF multimers in the circulation are thrombogenic, their activity in the circulation must be tightly regulated; thus, they are rapidly digested on release to smaller multimers (≤ 10 × 106 Da or 40 monomers19 ) via cleavage of the Y1605-M1606 bond in the A2 domain by ADAMTS13 (a disintegrin and metalloprotease with thrombospondin type 1 repeats, 13).20-22  Once in the bloodstream, VWF multimers circulate with a half-life of 8 to 12 hours. The mechanisms by which VWF is cleared are not yet fully understood, but recent evidence suggests a role for macrophages in the removal of the VWF/FVIII complex in both the liver and spleen.23 

Inherited quantitative and/or qualitative abnormalities in VWF are responsible for the bleeding disorder VWD. The prevalence of VWD, based on low VWF levels, may be up to 1%.24,25  However, the incidence of those requiring treatment is in the range of 0.002% to 0.01%.26  VWD is most commonly associated with mucosal hemorrhages, such as epistaxis, menorrhagia, and bleeding from the gums and gastrointestinal tract. Prolonged bleeding after trauma to skin or mucous membranes is also characteristic, especially during or after surgery. The most severely affected VWD patients have concomitant low levels of FVIII and are at risk of more severe and spontaneous deep tissue bleeds and hemarthroses.

The identification of more than 250 VWF gene mutations associated with all the types of VWD (database: www.sheffield.ac.uk/VWF) provides useful insights into the mechanisms underlying qualitative and quantitative VWF defects. However, the current clinical classification of VWD27,28  is not based on genetic defects but rather was designed for the purposes of optimizing treatment strategies (Table 1). Thus, a partial or complete quantitative deficiency of VWF results in so-called type 1 and type 3 VWD, respectively, whereas type 2 VWD is caused by qualitative defects.

Table 1

Phenotypic classification of VWD

VWD typePathophysiologic mechanism
Partial quantitative deficiency of VWF and/or FVIII 
2 Qualitative defects of VWF 
    A ↓ platelet-dependent VWF function, lack of HMWM 
    B ↑ platelet-dependent VWF function, lack of HMWM 
    M ↓ VWF function, normal multimers 
    N ↓ VWF binding to FVIII, normal multimers 
Severe/complete deficiency of VWF and moderately severe decrease of FVIII 
VWD typePathophysiologic mechanism
Partial quantitative deficiency of VWF and/or FVIII 
2 Qualitative defects of VWF 
    A ↓ platelet-dependent VWF function, lack of HMWM 
    B ↑ platelet-dependent VWF function, lack of HMWM 
    M ↓ VWF function, normal multimers 
    N ↓ VWF binding to FVIII, normal multimers 
Severe/complete deficiency of VWF and moderately severe decrease of FVIII 

Type 1 VWD is characterized by mild to severe quantitative defects of VWF (associated mostly with parallel decreases in FVIII levels) and accounts for 60% to 80% of all cases of VWD. Levels of VWF and FVIII are significantly lower in persons with blood group O; thus, the diagnosis of type 1 VWD must be made cautiously. Recent large studies in Europe and Canada have revealed that the majority of type 1 VWD is associated with mutations within VWF.29-31  The more severe forms of VWD with a high penetrance are associated with dominant, mostly single amino acid missense mutations in the D3 domain, that result in reduced intracellular transport of dimeric pro-VWF, or more rapid clearance of the protein from the circulation (for a review, see Nichols et al32 ).

Subtype 2A VWD, the most common qualitative variant, usually with an autosomal dominant inheritance pattern, is characterized by a decreased platelet-dependent function of VWF because of the loss of large and intermediate multimers. This is caused by mutations in either the propeptide or cystein knot domain, leading to defective assembly, and/or by mutations in the A2 domain that increase the susceptibility to proteolysis by ADAMTS13.33 

Subtype 2B VWD, usually inherited as an autosomal dominant trait, is associated with increased and often spontaneous binding of VWF to platelet GPIbα. The molecular defect resides within the A1 domain of VWF and is the result of various missense mutations.34  High molecular weight multimers are lost from plasma, as VWF binding to platelets on the one hand facilitates proteolysis by ADAMTS13, and on the other hand results in faster clearance of the platelet-VWF complexes. This gives rise to thrombocytopenia35  and explains the apparently paradoxical bleeding diathesis in these “gain-of-function” mutations.

Subtype 2M VWD is an autosomal dominant variant that displays decreased VWF function in the presence of a (near) normal multimeric distribution. Most of the associated mutations are within the A1 domain, causing impaired binding to GPIbα, without affecting multimer assembly.34  To date, one mutation in the VWF-A3 domain has been reported to result in defective VWF binding to collagen.36 

Subtype 2N VWD, an autosomal recessive disorder characterized by a decrease in the affinity of VWF for FVIII, is caused by approximately 20 homozygous or compound heterozygous missense mutations in the D′ or D3 FVIII binding domain. When there is a severe reduction in FVIII levels, type 2N VWD resembles mild or moderate hemophilia A.

Type 3 VWD is the most severe form of the disease, inherited in an autosomal recessive manner, with a prevalence ranging from 0.5 to 5 cases per million. It is caused by defective VWF synthesis, resulting in virtually complete absence of VWF, and a consequent severe reduction in circulating levels of FVIII. Almost 90 distinct mutations,34  associated with the type 3 phenotype, are scattered over the entire VWF gene and include partial or total deletions, small insertions, exon/intron boundary splice site mutations, which result in exon skipping or aberrant splicing, missense mutations, and frameshift mutations.

Acquired VWD is a rare syndrome that mimics the congenital form of VWD but occurs in persons with no personal or family history of bleeding. Several underlying diseases have been associated.37  These are most commonly myeloproliferative and lymphoproliferative disorders, where increased numbers of platelets may bind and clear the highest multimers. Autoimmune and cardiovascular disorders have also been associated. In situations with ultra-high shear stresses, particularly in stenosed areas, high multimer VWF cleavage by ADAMTS13 may be excessive, leading to reduced levels of active VWF.

In VWD patients who require treatment, the main goal is to replace the deficient or dysfunctional VWF protein, thereby restoring the hemostatic balance. At present, 2 primary strategies are used to stop spontaneous acute hemorrhages and to prevent major bleeding during invasive or surgical procedures: administration of desmopressin and infusion of plasma-derived VWF/FVIII concentrates. However, research continues, with the goal of developing safer and more effective preventative and therapeutic strategies. The use of interleukin-11, the introduction of standardized recombinant VWF preparations and advances in gene-based delivery approaches are some of the more promising alternatives undergoing active investigation, which may broaden the panel of future treatment modalities of VWD (Table 2).

Table 2

Overview of clinical and experimental VWD treatment strategies

Treatment statusAdvantagesDisadvantages
Current   
    Desmopressin Relatively inexpensive, no blood-borne contaminations, easy availability, self-administration Short-term effect, tachyphylaxis, variable response, mild side effects 
    VWF/FVIII concentrates Effective in nonresponders to DDAVP Short-term effect, higher costs, risk of contamination, variable quality 
Investigational   
    Interleukin-11 DDAVP surrogate/complement, sustained effect Minor to moderate side effects, increase in platelet count and fibrinogen levels 
Future   
    Recombinant VWF Standardized product, no risk of blood-borne contamination Short-term effect, no FVIII for acute needs 
    Gene therapy Potential of lifelong cure, increase in patient's quality of life Safety and ethical issues 
Treatment statusAdvantagesDisadvantages
Current   
    Desmopressin Relatively inexpensive, no blood-borne contaminations, easy availability, self-administration Short-term effect, tachyphylaxis, variable response, mild side effects 
    VWF/FVIII concentrates Effective in nonresponders to DDAVP Short-term effect, higher costs, risk of contamination, variable quality 
Investigational   
    Interleukin-11 DDAVP surrogate/complement, sustained effect Minor to moderate side effects, increase in platelet count and fibrinogen levels 
Future   
    Recombinant VWF Standardized product, no risk of blood-borne contamination Short-term effect, no FVIII for acute needs 
    Gene therapy Potential of lifelong cure, increase in patient's quality of life Safety and ethical issues 

Desmopressin

Desmopressin (1-deamino-8-D-arginine vasopressin [DDAVP]) is a synthetic analog of vasopressin that was found to raise VWF levels without the side effects of vasopressin.38,39  DDAVP induces exocytosis of Weibel-Palade bodies by binding to endothelial V2 receptors and activating cyclic adenosine monophosphate-mediated intracellular signaling pathways.40  An intravenous infusion of DDAVP (usually at a dose of 0.3 μg/kg diluted in 50 mL saline over 30 minutes) typically increases plasma levels of VWF 3- to 5-fold over baseline within the first hour after administration.

Concomitantly, DDAVP also increases plasma levels of FVIII. The precise mechanism of this rise is still poorly understood. The uncertainty relates particularly to the fact that the site of release of FVIII remains unclear, although Jacquemin et al suggest that lung microvascular endothelial cells might be an extrahepatic, DDAVP-releasable source.41  Moreover, when FVIII and VWF are expressed together in endothelial cells, VWF targets the FVIII to Weibel-Palade bodies, from which both can be secreted acutely in response to agonists.42 

Because high VWF levels are generally maintained for only 6 to 8 hours, DDAVP treatment must be repeated every 12 to 24 hours, depending on the type and the severity of the bleeding. Unfortunately, repeated use of desmopressin frequently results in a refractory response to subsequent doses.43 

Desmopressin administration is the treatment of choice for VWD type 1 because functionally intact VWF is present in endothelial storage granules of these patients.44,45  However, it is not effective in VWD type 3 or in severe cases of VWD type 1, yields a variable response in types 2A and 2M, and is contraindicated in patients with VWD type 2B because of the possibility of inducing transient thrombocytopenia. In view of the range in responses to DDAVP, a test infusion is recommended in all VWD patients (except type 2B). For poor responders, alternative treatments, such as plasma concentrates, rhIL-11, or rVWF may be considered. The variability in DDAVP response is not well explained, although several mechanisms have been proposed, including VWF mutations,46,47  causing differences in intracellular transport, secretion, and multimer composition of VWF48  and expression of A-antigen.49 

The main advantages of DDAVP are its low cost, ready availability, and suitability for self-administration. It is preferred over plasma concentrates, which carry the risk of transmission of blood-borne infections. Nonetheless, DDAVP may cause mild tachycardia, headache, facial flushing, as well as hyponatremia, particularly in children. Franchini et al also described an episode of deep vein thrombosis after DDAVP administration in a VWD patient undergoing orthopedic surgery,50  and, although not reported in patients with VWD, DDAVP has also been implicated in myocardial infarction and stroke.51,52 

VWF/FVIII concentrates

Current treatment of VWD patients who respond unsatisfactorily to DDAVP relies on transfusion therapy using VWF products manufactured from pooled human plasma. This furthermore is the only option for VWD type 3 patients because they lack a releasable pool of VWF. Because patients with VWD type 2 secrete a qualitatively abnormal VWF protein, DDAVP response is often minimal. In addition, patients in whom DDAVP is contraindicated, such as those with VWD type 2B, must be treated with plasma products. A wide variety of plasma concentrates is commercially available, but product composition differs significantly, depending on the purification scheme.53,54  Originally developed for hemophilia A, some products have a high proportion of FVIII (eg, Alphanate [antihemophilic factor]; Alpha Therapeutic, Los Angeles, CA), whereas other, newer formulations are essentially void of FVIII (eg, Wilfactin [VWF]; LFB, Les Ulis, France). Furthermore, the relative content and activity of VWF vary considerably, with many concentrates lacking high-molecular-weight VWF resulting from proteolysis during purification, which probably impacts on their overall effectiveness.53,54 

Because preparation of these concentrates requires the pooling of large quantities of human fresh-frozen plasma, untreated preparations that are/were unscreened and/or untreated carry a significant risk of transmitting blood-borne (viral) pathogens. Awareness of this problem became dramatically evident when it was recognized in the mid-late 1980s that up to 90% of severe hemophilia A patients had become seropositive for human immunodeficiency virus and hepatitis virus B and C after being treated with contaminated plasma products.55,56  Fortunately, during the last decades, major improvements have been made in the preparation of contaminant-free products. Using various techniques, including nanofiltration, solvent/detergent, and/or heat treatments, viral transmission has become less of an issue. Nevertheless, even adequately processed blood preparations are deemed to always carry a low but real risk of transmission of pathogens because current screening tests cannot exclude all pathogens. Nor can we fully anticipate what the future will hold, in terms of new blood transfusion-transmitted agents.57  It is therefore probable that plasma concentrates will never be entirely risk-free of transmitting infections, and alternative approaches are well worth pursuing.

Ideally, the concentrate of choice would be viral-inactivated and contain high amounts of clinically active, high-molecular-weight VWF multimers, as well as FVIII. Levels of FVIII will, however, be important to monitor, particularly in patients requiring repeated concentrate injections because sustained high FVIII levels may increase the risk of deep vein thrombosis.58  In that respect, it is important to appreciate also that the pharmacokinetics of FVIII levels after infusion of concentrates varies according to the preparation, and higher FVIII levels will be achieved more rapidly when FVIII is part of the concentrate. Thus, in type 3 VWD, hemostatic FVIII levels are reached only after 6 to 10 hours when concentrates lacking FVIII are used. Hence, this type of concentrate has limited utility in cases of acute, life-threatening bleeding episodes or emergency surgical procedures unless they are used in combination with exogenous recombinant FVIII. On the other hand, although VWF concentrates containing large quantities of FVIII may be preferred in emergency situations, their continued use in other situations is not recommended and maintenance with FVIII-deficient VWF concentrates (or recombinant VWF preparations) may be safer.

Clinical studies obtained over the last 20 years have demonstrated that currently used VWF/FVIII concentrates (most commonly Haemate P [antihemophilic factor]/Humate-P [antihemophilic factor]; ZLB Behring, Marburg, Germany) in patients with VWD are effective at stopping most bleeding episodes and preventing excessive blood loss during surgical procedures. When hemorrhage persists despite high VWF/FVIII levels, administration of platelet concentrates can be helpful, emphasizing the important role of platelet VWF in maintaining primary hemostasis.59-61  Platelet concentrates are effective, particularly in patients with type 3 VWD, probably because of their role in transporting VWF to sites of vascular injury where they secrete actively prothrombotic VWF.

Interleukin-11

Originally used to restore platelet counts after chemotherapy, recombinant human interleukin-11 (rhIL-11), a gp-130 signaling cytokine with hematopoietic and anti-inflammatory activity, was found to induce a gradual and sustained increase in VWF and FVIII levels in mice,62  dogs,63  and humans64  for as long as the drug was given. This is in contrast to DDAVP, which yields a rapid but transient increase in VWF/FVIII that becomes refractory to repeated infusions.

Side effects associated with rhIL-11 are generally mild or moderate and are easily managed. During recent phase 2 clinical trials, hypertension, fluid retention, and hypokalemia64,65  were reported as adverse events on infusion of rhIL-11 in patients with mild VWD. In cancer patients, the use of rhIL-11 has been reported to increase the susceptibility to transient atrial arrhythmias.66,67 

Interestingly, DDAVP administration during rhIL-11 treatment still results in the release of VWF from the Weibel-Palade bodies, suggesting that the mechanisms for VWF increase differ. These mechanisms have not yet been fully clarified, although IL-11 up-regulates VWF mRNA accumulation in dogs and humans, but not in mice.62  Nonetheless, the different mechanisms indicate that rhIL-11 could be used in combination with DDAVP to treat mild (quantitative) VWD, for example, to overcome DDAVP tachyphylaxis and in cases where prolonged high VWF levels are required. On the other hand, if proven safe in future clinical trials, the use of rhIL-11 will be of particular value for those patients who are refractory or unresponsive to DDAVP and/or who prefer a potentially safer treatment alternative to plasma-derived products.

In vitro–produced VWF

To circumvent the risks related to the use of blood as a source of therapeutic VWF, production of a VWF product by in vitro cell culture is an appealing alternative. Endothelial cell cultures, such as human umbilical vein endothelial cells and blood outgrowth endothelial cells (BOECs),68  appear to be good candidates because they have the natural capacity to correctly synthesize VWF. Yet, these primary human cell lines are not ideally suited for large-scale production of VWF. Interestingly, the cellular machinery responsible for correct assembly of VWF multimers is not unique to endothelial cells and megakaryocytes. VWF multimers can be properly formed in vitro by many cell types in low pH conditions.69  The propeptide itself seems to be the driving force for multimerization, possibly by providing noncovalent, intermolecular interactions between D1-D2 domains of different VWF subunits70  and by acting as an oxido-reductase during multimerization.71,72  As a consequence, efficient VWF biosynthesis has been observed in many different types of heterologous cells, including COS, CHO, 3T3, CV-1, AtT-20, RIN 5F, MDCKII, and HEK293 cells.73 

Indeed, a CHO cell–derived standardized recombinant VWF (rVWF) preparation for treatment of VWD is currently under development. This rVWF was initially produced in CHO cells coexpressing furin for proper propeptide removal.74  However, the protocol has been adapted, such that rVWF is coexpressed with recombinant FVIII in the cells. Propeptide cleavage occurs subsequently by exposing rVWF to recombinant CHO cell–derived furin. Preclinical studies showed that furin-processed rVWF has properties comparable with plasma-derived VWF in a variety of in vitro functional assays. One exception is that rVWF contains more intact multimers as it has not been exposed to ADAMTS13 processing.75,76  This apparently does not markedly alter the function of the VWF, at least in vitro. Pharmacokinetic properties of rVWF were investigated in murine and canine models of VWD where a slightly longer half-life for rVWF was observed in VWF knockout mice compared with plasma-derived VWF.75-77  Preliminary data show that the half-life of recombinant VWF can be further increased by PEGylation or by modifying the glycosylation pattern of VWF, although these modifications significantly reduce the functional activity of VWF.78,79  On the other hand, a prolonged half-life of VWF and concomitant increase of the half-life of coadministered FVIII might be beneficial for patients with hemophilia A.

Because pure rVWF preparations contain only VWF, concomitant administration of FVIII for VWD patients will be necessary for the treatment of acute bleeding and for the prevention of excessive bleeding during major surgery. Future clinical studies with rVWF will reveal its efficacy and, thus, its potential as an alternative to plasma VWF concentrates.

Gene therapy

Patients with severe bleeding phenotypes (VWD type 3 and some severe cases of types 1 and 2) not only have the looming threat of a serious hemorrhage but are also confronted with more general quality of life challenges that impact on education, work, social activities, and family life.80  Frequent hemarthroses often lead to arthropathies that limit the patient's mobility and independence.81,82  Recurrent epistaxis can be severe enough to cause anemia in children. Patients with frequent gastrointestinal or other bleeding episodes may require treatments every day or every other day, restricting work and social activities. Menorrhagia also has a negative impact on overall life activities.83,84  The daily management of symptoms, the unpredictable and potential life-threatening nature of bleeding episodes, and concerns about the future are furthermore associated with anxiety and depression.80  Overall, it is clear that treatment strategies that are preventative rather than reactive, with a sustained effectiveness, would be particularly valuable for these persons.

The benefits of long-term treatment of VWD can be deduced from the limited number of prophylaxis studies. Although long-term prophylaxis is not as common for VWD as for hemophilia A (probably because of an underestimation of the number of patients who would benefit85 ), recent data from Swedish surveys are encouraging.86,87  A substantial number of patients who received VWF concentrates over long periods of time exhibited fewer bleeding episodes, and all reported an improved quality of life. Furthermore, in children who began long-term prophylaxis before the age of 5, none developed arthropathy. Finally, in an Italian study, long-term prophylaxis stopped bleeding in VWD patients and reduced days in hospital.88 

Gene therapy for VWD offers the potential of a long-term, if not lifelong, correction of VWF deficiency, which would dramatically change the patient's personal comfort and quality of life. Instead of repetitive, on-demand replacement of the deficient/defective protein, a permanent correction of the underlying genetic defect would ideally cure the disorder. Indeed, VWD is a good candidate for gene therapy, as it is a monogenic disease. Moreover, VWF is secreted into the circulation; thus, organ- or tissue-specific targeting is not required. Although several safety and ethical issues remain to be resolved in the field of gene therapy, interest in using this approach for VWD is increasing.68,89-92 

In the past, the development of gene therapy for VWD was viewed as a daunting task, especially because of the large size of the VWF cDNA (8.4 kb), which impeded efficient incorporation into integrating viral vectors. Moreover, as mentioned earlier, the VWF protein undergoes several complex intracellular processing steps, all of which are required to yield a functional protein. Nonetheless, these challenges are being met. We recently demonstrated the feasibility of accommodating full-length VWF into a lentiviral vector (Figure 2).68  With this vector, expression of functional VWF could be completely restored in BOECs that were isolated from dogs with VWD type 3. It remains to be seen whether such genetically engineered autologous BOECs can be engrafted in vivo into VWD donors for long-term delivery of VWF. However, promising proof-of-principle experiments are available for the treatment of hemophilia A in animal models.93,94 

Figure 2

Expression of transgene-encoded VWF in VWD BOECs after lentiviral transduction. VWD BOECs were isolated from dogs with VWD type 3 and transduced with lentiviral vectors encoding full-length human VWF. VWF immunostaining revealed high concentrations of VWF present in both the cytoplasm and Weibel-Palade bodies (magnification) of transduced VWD BOECs (Lenti-CMV-huVWF) but not in VWD BOECs transduced with empty lentiviral particles (empty vector). Reprinted from De Meyer et al68  with permission.

Figure 2

Expression of transgene-encoded VWF in VWD BOECs after lentiviral transduction. VWD BOECs were isolated from dogs with VWD type 3 and transduced with lentiviral vectors encoding full-length human VWF. VWF immunostaining revealed high concentrations of VWF present in both the cytoplasm and Weibel-Palade bodies (magnification) of transduced VWD BOECs (Lenti-CMV-huVWF) but not in VWD BOECs transduced with empty lentiviral particles (empty vector). Reprinted from De Meyer et al68  with permission.

Close modal

With the availability of a lentiviral tool for VWF gene transfer, another interesting gene-based option to correct VWD would involve direct in vivo transduction after systemic vector administration. One drawback might be that lentiviral vectors predominantly target the liver; thus, the transgene expressing VWF would reside in liver cells. However, using the technique of hydrodynamic gene transfer in a murine model of severe VWD, we and others demonstrated that the liver indeed is capable of producing VWF containing the full range of multimers (Figure 3).90,92,95  Transgene-encoded, liver-expressed VWF could restore FVIII levels to normal and, most importantly, reversed the prolongation of the tail bleeding time.90,92,95  Furthermore, in a FeCl3-induced thrombosis model, there was restoration of the platelet plug-forming capacity, which is a major hemostatic function of VWF.92,96 

Figure 3

Multimer analysis of transgene encoded VWF expressed in the liver. Multimer analysis was performed on a plasma sample that was isolated from a VWD type 3 mouse 3 days after liver-directed hydrodynamic gene transfer of murine VWF cDNA (liver-expressed VWF). For comparison, the multimer pattern of plasma VWF from wild-type mice is shown (normal murine plasma [NMP]). Adapted from De Meyer et al92  with permission.

Figure 3

Multimer analysis of transgene encoded VWF expressed in the liver. Multimer analysis was performed on a plasma sample that was isolated from a VWD type 3 mouse 3 days after liver-directed hydrodynamic gene transfer of murine VWF cDNA (liver-expressed VWF). For comparison, the multimer pattern of plasma VWF from wild-type mice is shown (normal murine plasma [NMP]). Adapted from De Meyer et al92  with permission.

Close modal

When VWF is expressed in heterologous cells, one has to consider that proper digestion by ADAMTS13 might be impaired. Independent of the cellular source, circulating VWF will most probably be cleaved normally in the bloodstream. However, on secretion itself, specific VWF-endothelial interactions may facilitate digestion of UL-VWF, such as the anchoring of released VWF to the endothelial surface by αvβ397  and P-selectin.98  Hence, when VWF is expressed in nonendothelial cell types such as liver cells, efficient processing of VWF by ADAMTS13 could be hampered. Interestingly, however, in contrast to endothelial cells, liver cells do not seem to express UL-VWF multimers92  (and S.F.D.M. and K.V., unpublished data, September 2008). This could be related to differences in cellular machinery, such as the absence of a regulated secretory pathway in hepatocytes. Therefore, even in the absence of cleavage by ADAMTS13, liver-expressed VWF would not probably pose a thrombotic risk related to the circulation of UL-VWF multimers.

This is in accordance with the observed normal FeCl3-induced thrombus formation and the absence of spontaneous thrombotic complications in VWF knockout mice after gene transfer, even when liver-expressed VWF levels were 10-fold higher than in wild-type animals.92  Further studies using VWD animal models will provide more insight into the long-term therapeutic value of this liver-targeted approach.

These first steps toward gene therapy for VWD are only the tip of the iceberg. It is clear that, for each target disease, including severe VWD, a careful risk-benefit assessment of gene therapy and other therapeutic options will be required. With the current status of gene therapy, it is common sense that only the most severely affected patients (VWD type 3) would be considered for a gene-based treatment approach. Although VWD is not life-threatening and there are other available treatments, VWD, similar to hemophilia, is indeed an ideal candidate for gene therapy. However, in hemophilia, an increase of 1% to 2% in circulating levels of the deficient clotting factor can significantly modify the bleeding diathesis. Higher levels of VWF will most probably be needed to achieve hemostasis in VWD. Future studies, preferably in larger animal models of severe VWD such as dogs or pigs, will undoubtedly shed more light on this issue and further expand our knowledge on the possibilities and limitations of VWD gene therapy. As the field of gene therapy is making slow but steady progress, with promising results in a range of diseases, the tide of public opinion is shifting. Despite the isolated adverse events related to gene therapy, there is a large number of patients whose quality of life has been dramatically improved by having received gene therapy.99  Once issues on safety and efficiency are resolved, the appeal of treating severe VWD patients via gene therapy approaches will surely lead to its approval and wide use.

Anti-VWF alloantibodies

Alloantibodies directed against the substituted VWF may complicate the treatment of patients with VWD type 3, particularly in those patients with large VWF gene deletions.100  These antibodies may severely reduce the effectiveness of administered VWF, either by interfering with the functional domains of VWF or by reducing its half-life in the circulation. However, compared with FVIII, the development of inhibitory antibodies against VWF is quite rare (10%-15% of patients with VWD type 3), with only a few cases being described. Exceptionally, administration of VWF/FVIII concentrate to patients with VWF antibodies can induce life-threatening anaphylaxis.101  When such patients fail immune tolerization, on-demand treatment with recombinant FVIII or recombinant activated FVII helps to control bleeding. Antibody induction may also be limiting the applicability of gene therapy for VWD. Because VWF also is normally present in platelets, one could consider a gene therapy approach where VWF is only targeted to platelets, thereby minimizing exposure to the immune system. Such an approach has already been used successfully for FVIII in hemophilia A mice.102,103  However, whether platelet VWF alone would be sufficient for normal hemostasis is questionable, based on studies in chimeric pigs where platelet VWF alone did not correct the bleeding time or thrombus formation.104 

It is interesting to note that most of the current information on the biology and management of VWD is deduced from studies conducted in developed countries with a predominantly white population. Data on the epidemiology and management of VWD in developing countries are limited,105-107  although approximately 80% of the world's population lives in these countries. Whether the development of new treatments will add to better management of VWD patients in these countries is hard to predict. Efficient use of currently available treatments (DDAVP and plasma concentrates) in developing countries is hampered by budgetary issues and by lack of proper awareness and/or training in the management of bleeding disorders in general. Thus, whereas much less expensive medication for VWD would certainly help where VWD therapy is presently unaffordable, it may only reach patients in an environment of optimal awareness of the disorder. With the cost burden of plasma concentrates already being very high in developing countries, it is doubtful whether new treatments, such as the use of recombinant factors (IL-11 or VWF) or gene therapy, will help to reduce this burden.

In conclusion, more than 80 years have passed since Erik von Willebrand described the bleeding disorder that bears his name. In those early days, treatment strategies were far from adequate, and patients with severe forms of VWD were crippled before adulthood and had shortened life expectancy. Illustratively, Dr von Willebrand's first case, a 5-year-old girl, bled to death during her fourth menses at the age of 13. Over the past 50 years, our understanding of the bleeding disorder has steadily increased, resulting in dramatic improvements in the management of VWD. It will be exciting to unravel more aspects of VWF biology and to use this new knowledge in the optimization and design of treatments for VWD. Current strategies, using DDAVP or plasma concentrates, give satisfactory results, and alternative treatment options can further improve or at least broaden the panel of VWD treatment modalities. Whether the VWF molecule that comes to the rescue is endogenous, exogenous, recombinant, or even transgene encoded, it will always serve the same goal in VWD: to restore the functional deficiency of VWF and to rapidly and effectively correct the associated hemostatic abnormalities.

The authors thank Prof P. M. Mannucci (A. Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy) and Dr E. M. Conway (Katholieke Universiteit Leuven, Leuven, Belgium) for careful reading of the manuscript and helpful criticism.

S.F.D.M. and K.V. are postdoctoral fellows of the Fonds voor Wetenschappelijk Onderzoek, Vlaanderen, Belgium.

Contribution: S.F.D.M., H.D., and K.V. wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Simon F. De Meyer, Laboratory for Thrombosis Research, Interdisciplinary Research Center, KU Leuven Campus Kortrijk, E Sabbelaan 53, 8500 Kortrijk, Belgium; e-mail: simon.demeyer@kuleuven-kortrijk.be.

1
von Willebrand
 
EA
Hereditär pseudohemofili.
Fin Laekaresaellsk Hand
1926
, vol. 
68
 (pg. 
87
-
112
)
2
Nilsson
 
IM
Blomback
 
M
Von
 
FI
On an inherited autosomal hemorrhagic diathesis with antihemophilic globulin (AHG) deficiency and prolonged bleeding time.
Acta Med Scand
1957
, vol. 
159
 (pg. 
35
-
57
)
3
Nilsson
 
IM
Blomback
 
M
Jorpes
 
E
, et al. 
von Willebrand's disease and its correction with human plasma fraction 1-0.
Acta Med Scand
1957
, vol. 
159
 (pg. 
179
-
188
)
4
Sadler
 
JE
Shelton-Inloes
 
BB
Sorace
 
JM
, et al. 
Cloning and characterization of two cDNAs coding for human von Willebrand factor.
Proc Natl Acad Sci U S A
1985
, vol. 
82
 (pg. 
6394
-
6398
)
5
Verweij
 
CL
Diergaarde
 
PJ
Hart
 
M
Pannekoek
 
H
Full-length von Willebrand factor (vWF) cDNA encodes a highly repetitive protein considerably larger than the mature vWF subunit.
EMBO J
1986
, vol. 
5
 (pg. 
1839
-
1847
)
6
Emsley
 
J
Cruz
 
M
Handin
 
R
Liddington
 
R
Crystal structure of the von Willebrand Factor A1 domain and implications for the binding of platelet glycoprotein Ib.
J Biol Chem
1998
, vol. 
273
 (pg. 
10396
-
10401
)
7
Bienkowska
 
J
Cruz
 
M
Atiemo
 
A
Handin
 
R
Liddington
 
R
The von Willebrand factor A3 domain does not contain a metal ion-dependent adhesion site motif.
J Biol Chem
1997
, vol. 
272
 (pg. 
25162
-
25167
)
8
Lankhof
 
H
van Hoeij
 
M
Schiphorst
 
ME
, et al. 
A3 domain is essential for interaction of von Willebrand factor with collagen type III.
Thromb Haemost
1996
, vol. 
75
 (pg. 
950
-
958
)
9
Sixma
 
JJ
Schiphorst
 
ME
Verweij
 
CL
Pannekoek
 
H
Effect of deletion of the A1 domain of von Willebrand factor on its binding to heparin, collagen and platelets in the presence of ristocetin.
Eur J Biochem
1991
, vol. 
196
 (pg. 
369
-
375
)
10
Martin
 
C
Morales
 
LD
Cruz
 
MA
Purified A2 domain of von Willebrand factor binds to the active conformation of von Willebrand factor and blocks the interaction with platelet glycoprotein Ibalpha.
J Thromb Haemost
2007
, vol. 
5
 (pg. 
1363
-
1370
)
11
Nishio
 
K
Anderson
 
PJ
Zheng
 
XL
Sadler
 
JE
Binding of platelet glycoprotein Ib alpha to von Willebrand factor domain A1 stimulates the cleavage of the adjacent domain A2 by ADAMTS13.
Proc Natl Acad Sci U S A
2004
, vol. 
101
 (pg. 
10578
-
10583
)
12
Ulrichts
 
H
Udvardy
 
M
Lenting
 
PJ
, et al. 
Shielding of the A1 domain by the D′D3 domains of von Willebrand factor modulates its interaction with platelet glycoprotein Ib-IX-V.
J Biol Chem
2006
, vol. 
281
 (pg. 
4699
-
4707
)
13
Savage
 
B
Saldivar
 
E
Ruggeri
 
ZM
Initiation of platelet adhesion by arrest onto fibrinogen or translocation on von Willebrand factor.
Cell
1996
, vol. 
84
 (pg. 
289
-
297
)
14
Doggett
 
TA
Girdhar
 
G
Lawshe
 
A
, et al. 
Selectin-like kinetics and biomechanics promote rapid platelet adhesion in flow: the GPIb(alpha)-vWF tether bond.
Biophys J
2002
, vol. 
83
 (pg. 
194
-
205
)
15
Mody
 
NA
Lomakin
 
O
Doggett
 
TA
Diacovo
 
TG
King
 
MR
Mechanics of transient platelet adhesion to von Willebrand factor under flow.
Biophys J
2005
, vol. 
88
 (pg. 
1432
-
1443
)
16
Yago
 
T
Lou
 
J
Wu
 
T
, et al. 
Platelet glycoprotein Ibalpha forms catch bonds with human WT vWF but not with type 2B von Willebrand disease vWF.
J Clin Invest
2008
, vol. 
118
 (pg. 
3195
-
3207
)
17
Yago
 
T
Lou
 
J
Wu
 
T
, et al. 
Platelet glycoprotein lb alpha forms catch bonds with human WT vWF but not with type 2B von Willebrand disease vWF.
J Clin Invest
2008
, vol. 
118
 (pg. 
3195
-
3207
)
18
Giblin
 
JP
Hewlett
 
LJ
Hannah
 
MJ
Basal secretion of von Willebrand factor from human endothelial cells.
Blood
2008
, vol. 
112
 (pg. 
957
-
964
)
19
Fowler
 
WE
Fretto
 
LJ
Hamilton
 
KK
Erickson
 
HP
McKee
 
PA
Substructure of human von Willebrand factor.
J Clin Invest
1985
, vol. 
76
 (pg. 
1491
-
1500
)
20
Fujikawa
 
K
Suzuki
 
H
McMullen
 
B
Chung
 
D
Purification of human von Willebrand factor-cleaving protease and its identification as a new member of the metalloproteinase family.
Blood
2001
, vol. 
98
 (pg. 
1662
-
1666
)
21
Gerritsen
 
HE
Robles
 
R
Lammle
 
B
Furlan
 
M
Partial amino acid sequence of purified von Willebrand factor-cleaving protease.
Blood
2001
, vol. 
98
 (pg. 
1654
-
1661
)
22
Dent
 
JA
Berkowitz
 
SD
Ware
 
J
Kasper
 
CK
Ruggeri
 
ZM
Identification of a cleavage site directing the immunochemical detection of molecular abnormalities in type IIA von Willebrand factor.
Proc Natl Acad Sci U S A
1990
, vol. 
87
 (pg. 
6306
-
6310
)
23
van Schooten
 
CJ
Shahbazi
 
S
Groot
 
E
, et al. 
Macrophages contribute to the cellular uptake of von Willebrand factor and factor VIII in vivo.
Blood
2008
, vol. 
112
 (pg. 
1704
-
1712
)
24
Rodeghiero
 
F
Castaman
 
G
Dini
 
E
Epidemiological investigation of the prevalence of von Willebrand's disease.
Blood
1987
, vol. 
69
 (pg. 
454
-
459
)
25
Werner
 
EJ
Broxson
 
EH
Tucker
 
EL
, et al. 
Prevalence of von Willebrand disease in children: a multiethnic study.
J Pediatr
1993
, vol. 
123
 (pg. 
893
-
898
)
26
Sadler
 
JE
Mannucci
 
PM
Berntorp
 
E
, et al. 
Impact, diagnosis and treatment of von Willebrand disease.
Thromb Haemost
2000
, vol. 
84
 (pg. 
160
-
174
)
27
Sadler
 
JE
A revised classification of von Willebrand disease: for the Subcommittee on von Willebrand Factor of the Scientific and Standardization Committee of the International Society on Thrombosis and Haemostasis.
Thromb Haemost
1994
, vol. 
71
 (pg. 
520
-
525
)
28
Sadler
 
JE
Budde
 
U
Eikenboom
 
JC
, et al. 
Update on the pathophysiology and classification of von Willebrand disease: a report of the Subcommittee on von Willebrand Factor.
J Thromb Haemost
2006
, vol. 
4
 (pg. 
2103
-
2114
)
29
Eikenboom
 
J
Van Marion
 
V
Putter
 
H
, et al. 
Linkage analysis in families diagnosed with type 1 von Willebrand disease in the European study, molecular and clinical markers for the diagnosis and management of type 1 VWD.
J Thromb Haemost
2006
, vol. 
4
 (pg. 
774
-
782
)
30
James
 
PD
Notley
 
C
Hegadorn
 
C
, et al. 
The mutational spectrum of type 1 von Willebrand disease: results from a Canadian cohort study.
Blood
2007
, vol. 
109
 (pg. 
145
-
154
)
31
Goodeve
 
A
Genetics of type 1 von Willebrand disease.
Curr Opin Hematol
2007
, vol. 
14
 (pg. 
444
-
449
)
32
Nichols
 
WL
Hultin
 
MB
James
 
AH
, et al. 
von Willebrand disease (VWD): evidence-based diagnosis and management guidelines, the National Heart, Lung, and Blood Institute (NHLBI) Expert Panel report (USA).
Haemophilia
2008
, vol. 
14
 (pg. 
171
-
232
)
33
O'Brien
 
LA
Sutherland
 
JJ
Weaver
 
DF
Lillicrap
 
D
Theoretical structural explanation for Group I, Group II, type 2A von Willebrand disease mutations.
J Thromb Haemost
2005
, vol. 
3
 (pg. 
796
-
797
)
34
Lillicrap
 
D
von Willebrand disease—phenotype versus genotype: deficiency versus disease.
Thromb Res
2007
, vol. 
120
 
suppl 1
(pg. 
S11
-
S16
)
35
Federici
 
AB
Mannucci
 
PM
Castaman
 
G
, et al. 
Clinical and molecular predictors of thrombocytopenia and risk of bleeding in patients with von Willebrand disease type 2B: a cohort study of 67 patients.
Blood
2009
, vol. 
113
 (pg. 
526
-
534
)
36
Ribba
 
AS
Loisel
 
I
Lavergne
 
JM
, et al. 
Ser968Thr mutation within the A3 domain of von Willebrand factor (VWF) in two related patients leads to a defective binding of VWF to collagen.
Thromb Haemost
2001
, vol. 
86
 (pg. 
848
-
854
)
37
Federici
 
AB
Rand
 
JH
Mannucci
 
PM
Acquired von Willebrand syndrome: an important bleeding complication to be considered in patients with lymphoproliferative and myeloproliferative disorders.
Hematol J
2001
, vol. 
2
 (pg. 
358
-
362
)
38
Cash
 
JD
Gader
 
AM
da Costa
 
J
Br J Haematol
1974
, vol. 
27
 
Proceedings: the release of plasminogen activator and factor VIII to lysine vasopressin, arginine vasopressin, I-desamino-8-d-arginine vasopressin, angiotensin and oxytocin in man
(pg. 
363
-
364
)
39
Mannucci
 
PM
Aberg
 
M
Nilsson
 
IM
Robertson
 
B
Mechanism of plasminogen activator and factor VIII increase after vasoactive drugs.
Br J Haematol
1975
, vol. 
30
 (pg. 
81
-
93
)
40
Kaufmann
 
JE
Vischer
 
UM
Cellular mechanisms of the hemostatic effects of desmopressin (DDAVP).
J Thromb Haemost
2003
, vol. 
1
 (pg. 
682
-
689
)
41
Jacquemin
 
M
Neyrinck
 
A
Hermanns
 
MI
, et al. 
FVIII production by human lung microvascular endothelial cells.
Blood
2006
, vol. 
108
 (pg. 
515
-
517
)
42
Haberichter
 
SL
Shi
 
Q
Montgomery
 
RR
Regulated release of VWF and FVIII and the biologic implications.
Pediatr Blood Cancer
2006
, vol. 
46
 (pg. 
547
-
553
)
43
Mannucci
 
PM
Bettega
 
D
Cattaneo
 
M
Patterns of development of tachyphylaxis in patients with haemophilia and von Willebrand disease after repeated doses of desmopressin (DDAVP).
Br J Haematol
1992
, vol. 
82
 (pg. 
87
-
93
)
44
Mannucci
 
PM
Lombardi
 
R
Bader
 
R
, et al. 
Heterogeneity of type I von Willebrand disease: evidence for a subgroup with an abnormal von Willebrand factor.
Blood
1985
, vol. 
66
 (pg. 
796
-
802
)
45
Rodeghiero
 
F
Castaman
 
G
Di Bona
 
E
, et al. 
Hyper-responsiveness to DDAVP for patients with type I von Willebrand's disease and normal intra-platelet von Willebrand factor.
Eur J Haematol
1988
, vol. 
40
 (pg. 
163
-
167
)
46
Castaman
 
G
Lethagen
 
S
Federici
 
AB
, et al. 
Response to desmopressin is influenced by the genotype and phenotype in type 1 von Willebrand disease (VWD): results from the European Study MCMDM-1VWD.
Blood
2008
, vol. 
111
 (pg. 
3531
-
3539
)
47
Millar
 
CM
Riddell
 
AF
Brown
 
SA
, et al. 
Survival of von Willebrand factor released following DDAVP in a type 1 von Willebrand disease cohort: influence of glycosylation, proteolysis and gene mutations.
Thromb Haemost
2008
, vol. 
99
 (pg. 
916
-
924
)
48
Ruggeri
 
ZM
Mannucci
 
PM
Lombardi
 
R
Federici
 
AB
Zimmerman
 
TS
Multimeric composition of factor VIII/von Willebrand factor following administration of DDAVP: implications for pathophysiology and therapy of von Willebrand's disease subtypes.
Blood
1982
, vol. 
59
 (pg. 
1272
-
1278
)
49
Brown
 
SA
Collins
 
PW
Bowen
 
DJ
Heterogeneous detection of A-antigen on von Willebrand factor derived from platelets, endothelial cells and plasma.
Thromb Haemost
2002
, vol. 
87
 (pg. 
990
-
996
)
50
Franchini
 
M
Krampera
 
M
Veneri
 
D
Deep vein thrombosis after orthopedic surgery in a patient with type 1 von Willebrand disease and mutations in the MTHFR and beta-fibrinogen genes.
Thromb Haemost
2003
, vol. 
90
 (pg. 
963
-
964
)
51
Bond
 
L
Bevan
 
D
Myocardial infarction in a patient with hemophilia treated with DDAVP.
N Engl J Med
1988
, vol. 
318
 pg. 
121
 
52
Byrnes
 
JJ
Larcada
 
A
Moake
 
JL
Thrombosis following desmopressin for uremic bleeding.
Am J Hematol
1988
, vol. 
28
 (pg. 
63
-
65
)
53
Budde
 
U
Metzner
 
HJ
Muller
 
HG
Comparative analysis and classification of von Willebrand factor/factor VIII concentrates: impact on treatment of patients with von Willebrand disease.
Semin Thromb Hemost
2006
, vol. 
32
 (pg. 
626
-
635
)
54
Mazurier
 
C
Composition, quality control, and labeling of plasma-derived products for the treatment of von Willebrand disease.
Semin Thromb Hemost
2006
, vol. 
32
 (pg. 
529
-
536
)
55
Kroner
 
BL
Rosenberg
 
PS
Aledort
 
LM
Alvord
 
WG
Goedert
 
JJ
HIV-1 infection incidence among persons with hemophilia in the United States and western Europe, 1978-1990: Multicenter Hemophilia Cohort Study.
J Acquir Immune Defic Syndr
1994
, vol. 
7
 (pg. 
279
-
286
)
56
Troisi
 
CL
Hollinger
 
FB
Hoots
 
WK
, et al. 
A multicenter study of viral-hepatitis in a United-States hemophilic population.
Blood
1993
, vol. 
81
 (pg. 
412
-
418
)
57
Klein
 
HG
Pathogen inactivation technology: cleansing the blood supply.
J Intern Med
2005
, vol. 
257
 (pg. 
224
-
237
)
58
Mannucci
 
PM
Venous thromboembolism in von Willebrand disease.
Thromb Haemost
2002
, vol. 
88
 (pg. 
378
-
379
)
59
Castillo
 
R
Monteagudo
 
J
Escolar
 
G
, et al. 
Hemostatic effect of normal platelet transfusion in severe von Willebrand disease patients.
Blood
1991
, vol. 
77
 (pg. 
1901
-
1905
)
60
Boda
 
Z
Pfliegler
 
G
Harsfalvi
 
J
Rak
 
K
Treatment of the severe bleeding episode in type III von Willebrand's disease by simultaneous administration of cryoprecipitate and platelet concentrate.
Blood Coagul Fibrinolysis
1991
, vol. 
2
 (pg. 
775
-
777
)
61
Castillo
 
R
Escolar
 
G
Monteagudo
 
J
, et al. 
Hemostasis in patients with severe von Willebrand disease improves after normal platelet transfusion and normalizes with further correction of the plasma defect.
Transfusion
1997
, vol. 
37
 (pg. 
785
-
790
)
62
Denis
 
CV
Kwack
 
K
Saffaripour
 
S
, et al. 
Interleukin 11 significantly increases plasma von Willebrand factor and factor VIII in wild type and von Willebrand disease mouse models.
Blood
2001
, vol. 
97
 (pg. 
465
-
472
)
63
Olsen
 
EH
McCain
 
AS
Merricks
 
EP
, et al. 
Comparative response of plasma VWF in dogs to up-regulation of VWF mRNA by interleukin-11 versus Weibel-Palade body release by desmopressin (DDAVP).
Blood
2003
, vol. 
102
 (pg. 
436
-
441
)
64
Ragni
 
MV
Jankowitz
 
RC
Chapman
 
HL
, et al. 
A phase II prospective open-label escalating dose trial of recombinant interleukin-11 in mild von Willebrand disease.
Haemophilia
2008
, vol. 
14
 (pg. 
968
-
977
)
65
Jankowitz
 
RC
Ragni
 
MV
Chapman
 
HL
, et al. 
Recombinant interleukin-11 (rhIL-11) in women with refractory menorrhagia and von Willebrand disease [abstract].
Blood
2008
, vol. 
112
  
Abstract 1210
66
Gordon
 
MS
Caskill-Stevens
 
WJ
Battiato
 
LA
, et al. 
A phase I trial of recombinant human interleukin-11 (neumega rhIL-11 growth factor) in women with breast cancer receiving chemotherapy.
Blood
1996
, vol. 
87
 (pg. 
3615
-
3624
)
67
Tepler
 
I
Elias
 
L
Smith
 
JW
, et al. 
A randomized placebo-controlled trial of recombinant human interleukin-11 in cancer patients with severe thrombocytopenia due to chemotherapy.
Blood
1996
, vol. 
87
 (pg. 
3607
-
3614
)
68
De Meyer
 
SF
Vanhoorelbeke
 
K
Chuah
 
MK
, et al. 
Phenotypic correction of von Willebrand disease type 3 blood-derived endothelial cells with lentiviral vectors expressing von Willebrand factor.
Blood
2006
, vol. 
107
 (pg. 
4728
-
4736
)
69
Mayadas
 
TN
Wagner
 
DD
In vitro multimerization of von Willebrand factor is triggered by low pH: importance of the propolypeptide and free sulfhydryls.
J Biol Chem
1989
, vol. 
264
 (pg. 
13497
-
13503
)
70
Verweij
 
CL
Hart
 
M
Pannekoek
 
H
Proteolytic cleavage of the precursor of von Willebrand factor is not essential for multimer formation.
J Biol Chem
1988
, vol. 
263
 (pg. 
7921
-
7924
)
71
Mayadas
 
TN
Wagner
 
DD
Vicinal cysteines in the prosequence play a role in von Willebrand factor multimer assembly.
Proc Natl Acad Sci U S A
1992
, vol. 
89
 (pg. 
3531
-
3535
)
72
Purvis
 
AR
Gross
 
J
Dang
 
LT
, et al. 
Two Cys residues essential for von Willebrand factor multimer assembly in the Golgi.
Proc Natl Acad Sci U S A
2007
, vol. 
104
 (pg. 
15647
-
15652
)
73
Hannah
 
MJ
Williams
 
R
Kaur
 
J
Hewlett
 
LJ
Cutler
 
DF
Biogenesis of Weibel-Palade bodies.
Semin Cell Dev Biol
2002
, vol. 
13
 (pg. 
313
-
324
)
74
Plaimauer
 
B
Schlokat
 
U
Turecek
 
PL
, et al. 
Recombinant von Willebrand factor: preclinical development.
Semin Thromb Hemost
2001
, vol. 
27
 (pg. 
395
-
403
)
75
Turecek
 
PL
Mitterer
 
A
Mathiessen
 
HP
, et al. 
Downstream processing and characterization of a novel recombinant von Willebrand factor product:
2008
Proceedings of the European Workshop on von Willebrand Factor and von Willebrand Disease
Antwerp, Belgium
pg. 
167
 
76
Turecek
 
PL
Matthiessen
 
HP
Mitterer
 
A
, et al. 
Biochemical and functional characterization of a serum-free rVWF drug candidate [abstract].
Blood
2006
, vol. 
108
  
Abstract 1017
77
Nichols
 
TC
Merricks
 
E
Muchitsch
 
EM
, et al. 
Pharmacokinetics of rVWF in dogs and mice with severe VWD [abstract].
J Thromb Haemost
2007
, vol. 
5
 
suppl 2
 
Abstract P-W-197
78
Turecek
 
PL
Scheiflinger
 
F
Siekmann
 
J
, et al. 
Biochemical and functional characterization of PEGylated rVWF [abstract].
Blood
2006
, vol. 
108
  
Abstract 1021
79
Turecek
 
PL
Siekmann
 
J
Weber
 
A
, et al. 
Modification of rVWF with polysialic acid: biochemical and functional characterization in mice with VWD [abstract].
Blood
2006
, vol. 
108
  
Abstract 1001
80
Barlow
 
JH
Stapley
 
J
Ellard
 
DR
Living with haemophilia and von Willebrand's: a descriptive qualitative study.
Patient Educ Couns
2007
, vol. 
68
 (pg. 
235
-
242
)
81
Lak
 
M
Peyvandi
 
F
Mannucci
 
PM
Clinical manifestations and complications of childbirth and replacement therapy in 385 Iranian patients with type 3 von Willebrand disease.
Br J Haematol
2000
, vol. 
111
 (pg. 
1236
-
1239
)
82
Federici
 
AB
Clinical diagnosis of von Willebrand disease.
Haemophilia
2004
, vol. 
10
 
suppl 4
(pg. 
169
-
176
)
83
Rodeghiero
 
F
Management of menorrhagia in women with inherited bleeding disorders: general principles and use of desmopressin.
Haemophilia
2008
, vol. 
14
 (pg. 
21
-
30
)
84
Kirtava
 
A
Drews
 
C
Lally
 
C
Dilley
 
A
Evatt
 
B
Medical, reproductive and psychosocial experiences of women diagnosed with von Willebrand's disease receiving care in haemophilia treatment centres: a case-control study.
Haemophilia
2003
, vol. 
9
 (pg. 
292
-
297
)
85
Berntorp
 
E
Prophylaxis in von Willebrand disease.
Haemophilia
2008
, vol. 
14
 
suppl 5
(pg. 
47
-
53
)
86
Lethagen
 
S
Clinical experience of prophylactic treatment in von Willebrand disease.
Thromb Res
2006
, vol. 
118
 
suppl 1
(pg. 
S9
-
S11
)
87
Berntorp
 
E
Petrini
 
P
Long-term prophylaxis in von Willebrand disease.
Blood Coagul Fibrinolysis
2005
, vol. 
16
 
suppl 1
(pg. 
S23
-
S26
)
88
Federici
 
AB
Gianniello
 
F
Canciani
 
MT
Mannucci
 
PM
Secondary long-term prophylaxis in severe patients with von Willebrand's disease: an Italian cohort study [abstract].
Blood
2005
, vol. 
106
  
Abstract 1782
89
De Meyer
 
SF
Pareyn
 
I
Baert
 
J
Deckmyn
 
H
Vanhoorelbeke
 
K
False positive results in chimeraplasty for von Willebrand disease.
Thromb Res
2007
, vol. 
119
 (pg. 
93
-
104
)
90
Lenting
 
PJ
de Groot
 
PG
De Meyer
 
SF
, et al. 
Correction of the bleeding time in von Willebrand factor (VWF)-deficient mice using murine VWF.
Blood
2007
, vol. 
109
 (pg. 
2267
-
2268
)
91
Pergolizzi
 
RG
Jin
 
GC
Chan
 
D
, et al. 
Correction of a murine model of von Willebrand disease by gene transfer.
Blood
2006
, vol. 
108
 (pg. 
862
-
869
)
92
De Meyer
 
SF
Vandeputte
 
N
Pareyn
 
I
, et al. 
Restoration of plasma von Willebrand factor deficiency is sufficient to correct thrombus formation after gene therapy for severe von Willebrand disease.
Arterioscler Thromb Vasc Biol
2008
, vol. 
28
 (pg. 
1621
-
1626
)
93
Lin
 
Y
Chang
 
L
Solovey
 
A
, et al. 
Use of blood outgrowth endothelial cells for gene therapy for hemophilia A.
Blood
2002
, vol. 
99
 (pg. 
457
-
462
)
94
Matsui
 
H
Shibata
 
M
Brown
 
B
, et al. 
Ex vivo gene therapy for hemophilia A that enhances safe delivery and sustained in vivo factor VIII expression from lentivirally engineered endothelial progenitors.
Stem Cells
2007
, vol. 
25
 (pg. 
2660
-
2669
)
95
Marx
 
I
Lenting
 
PJ
Adler
 
T
, et al. 
Correction of bleeding symptoms in von Willebrand factor-deficient mice by liver-expressed von Willebrand factor mutants.
Arterioscler Thromb Vasc Biol
2008
, vol. 
28
 (pg. 
419
-
424
)
96
Marx
 
I
Christophe
 
OD
Lenting
 
PJ
, et al. 
Altered thrombus formation in von Willebrand factor-deficient mice expressing von Willebrand factor variants with defective binding to collagen or GPIIbIIIa.
Blood
2008
, vol. 
112
 (pg. 
603
-
609
)
97
Huang
 
J
Roth
 
R
Heuser
 
JE
Sadler
 
JE
Integrin alpha (v) beta(3) on human endothelial cells binds von Willebrand factor strings under fluid shear stress.
Blood
2009
, vol. 
113
 (pg. 
1589
-
1597
)
98
Padilla
 
A
Moake
 
JL
Bernardo
 
A
, et al. 
P-selectin anchors newly released ultralarge von Willebrand factor multimers to the endothelial cell surface.
Blood
2004
, vol. 
103
 (pg. 
2150
-
2156
)
99
Edelstein
 
ML
Abedi
 
MR
Wixon
 
J
Gene therapy clinical trials worldwide to 2007: an update.
J Gene Med
2007
, vol. 
9
 (pg. 
833
-
842
)
100
Shelton-Inloes
 
BB
Chehab
 
FF
Mannucci
 
PM
Federici
 
AB
Sadler
 
JE
Gene deletions correlate with the development of alloantibodies in von Willebrand disease.
J Clin Invest
1987
, vol. 
79
 (pg. 
1459
-
1465
)
101
Mannucci
 
PM
Tamaro
 
G
Narchi
 
G
, et al. 
Life-threatening reaction to factor VIII concentrate in a patient with severe von Willebrand disease and alloantibodies to von Willebrand factor.
Eur J Haematol
1987
, vol. 
39
 (pg. 
467
-
470
)
102
Shi
 
Q
Wilcox
 
DA
Fahs
 
SA
, et al. 
Lentivirus-mediated platelet-derived factor VIII gene therapy in murine haemophilia A.
J Thromb Haemost
2007
, vol. 
5
 (pg. 
352
-
361
)
103
Shi
 
Q
Fahs
 
SA
Wilcox
 
DA
, et al. 
Syngeneic transplantation of hematopoietic stem cells that are genetically modified to express factor VIII in platelets restores hemostasis to hemophilia A mice with preexisting FVIII immunity.
Blood
2008
, vol. 
112
 (pg. 
2713
-
2721
)
104
Nichols
 
TC
Samama
 
CM
Bellinger
 
DA
, et al. 
Function of von Willebrand factor after crossed bone marrow transplantation between normal and von Willebrand disease pigs: effect on arterial thrombosis in chimeras.
Proc Natl Acad Sci U S A
1995
, vol. 
92
 (pg. 
2455
-
2459
)
105
Mannucci
 
PM
Management of von Willebrand disease in developing countries.
Semin Thromb Hemost
2005
, vol. 
31
 (pg. 
602
-
609
)
106
Srivastava
 
A
von Willebrand disease in the developing world.
Semin Hematol
2005
, vol. 
42
 (pg. 
36
-
41
)
107
Srivastava
 
A
Rodeghiero
 
F
Epidemiology of von Willebrand disease in developing countries.
Semin Thromb Hemost
2005
, vol. 
31
 (pg. 
569
-
576
)
Sign in via your Institution