Non-Classical Anti-Factor VIII C2 Domain Antibodies Are Pathogenic in a Murine in Vivo Bleeding Model.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2027-2027
Author(s):  
Shannon Meeks ◽  
John F. Healey ◽  
Ernest T. Parker ◽  
Rachel T. Barrow ◽  
John (Pete) Lollar

Abstract Approximately 30% of patients with severe hemophilia A will develop inhibitory antibodies to factor VIII (fVIII inhibitors). In addition, autoimmune antibodies to fVIII can develop in non-hemophiliacs, producing acquired hemophilia A, which frequently produces life- or limb-threatening bleeding. Patients with congenital hemophilia who develop inhibitors usually have a polyclonal antibody response directed against the A2 and C2 domains of fVIII. Patients with acquired hemophilia typically have a more limited B-cell epitope response with antibodies directed against the A2 or C2 domain not both. Classical anti-C2 antibodies inhibit the binding of fVIII to phospholipid membranes and to von Willebrand factor. We recently have identified anti-C2 antibodies that inhibit the activation of fVIII, but do not inhibit the binding of fVIII to phospholipid membranes or to von Willebrand factor. These non-classical inhibitors are found in the plasmas of most inhibitor patients (Meeks, S.L. et al. Blood112, 1151-1153, 2008). The pathogenicity of classical and non-classical murine anti-human fVIII monoclonal antibodies (MAbs) was tested in a murine in vivo bleeding model. MAbs were injected into the tail veins of –hemophilia A mice to a peak plasma concentration of 60 nM followed by an injection human B domain-deleted fVIII to a concentration of 2 nM. At 2 hours the mice were anesthetized and a 4 mm tail snip was made. The amount of blood lost into a collection tube of normal saline over 40 minutes was measured. 4A4 is a type I anti-A2 inhibitor with an inhibitory titer of 40,000 Bethesda units (BU)/mg IgG. I54 and 1B5 are classical type I anti-C2 inhibitors with inhibitory titers of 1300 and 930 BU/mg IgG, respectively. 2-77 is a non-classical type II anti-C2 inhibitor that produces a residual fVIII level of 40% at saturating concentrations and whose titer is 21,000 BU/mg IgG. 2-117 is a non-classical anti-C2 MAb with inhibitory activity less than 0.4 BU/mg IgG. All of these MAbs except 2-117 significantly increased blood loss over control mice injected with fVIII alone (p= 0.01-0.02, Mann-Whitney Test) (Fig .1). The amount of blood loss was similar at these saturating concentrations of antibody despite inhibitory titers ranging from 930-40,000 BU/mg IgG. Increasing the dose of fVIII to 4 nM could overcome the bleeding diathesis produced by the non-classical MAb 2-77, but not the type I antibodies, 4A4 and I54. Similar results were seen in the in vitro Bethesda assay where 4A4 completely inhibited both 1 U/ml and 3 U/ml fVIII at saturating concentrations, while 2-77 had 40% residual activity with either 1 or 3 U/ml fVIII (0.4 U and 1.2 U respectively) (Fig. 2). These results suggest that high-dose fVIII rather than bypassing agents may be warranted in patients with an inhibitor response dominated by non-classical anti-C2 antibodies. FIGURE 1. FIGURE 1. FIGURE 2. FIGURE 2.

Blood ◽  
1996 ◽  
Vol 87 (5) ◽  
pp. 1809-1816 ◽  
Author(s):  
AJ Vlot ◽  
SJ Koppelman ◽  
JC Meijers ◽  
C Dama ◽  
HM van den Berg ◽  
...  

The binding of factor VIII to von Willebrand factor (vWF) is essential for the protection of factor VIII against proteolytic degradation in plasma. We have characterized the binding kinetics of human factor VIII with vWF using a centrifugation binding assay. Purified or plasma vWF was immobilized with a monoclonal antibody (MoAb RU1) covalently linked to Sepharose (Pharmacia LKB Biotechnology, Uppsala, Sweden). Factor VIII was incubated with vWF-RU1-Sepharose and unbound factor VIII was separated from bound factor VIII by centrifugation. The amount of bound factor VIII was determined from the decrease of factor VIII activity in the supernatant. Factor VIII binding to vWF-RU1-Sepharose conformed to the Langmuir model for independent binding sites with a Kd of 0.46 +/- 0.12 nmol/L, and a stoichiometry of 1.3 factor VIII molecules per vWF monomer at saturation, suggesting that each vWF subunit contains a binding site for factor VIII. Competition experiments were performed with a recombinant vWF (deltaA2-rvWF), lacking residues 730 to 910 which contain the epitope for MoAB RU1. DeltaA2-rvWF effectively displaced previously bound factor VIII, confirming that factor VIII binding to vWF-RU1-Sepharose was reversible. To determine the association rate constant (k(on)) and the dissociation rate constant (k(off)), factor VIII was incubated with vWF-RU1-Sepharose for various time intervals. The observed association kinetics conformed to a simple bimolecular association reaction with k(on) = 5.9 +/- 1.9 x 10(6) M(-1) s(-1) and k(off) = 1.6 +/- 1.2 x 10(-3) s(-1) (mean +/- SD). Similar values were obtained from the dissociation kinetics measured after dilution of preformed factor VIII-vWF-RU1-Sepharose complexes. Identical rate constants were obtained for factor VIII binding to vWF from normal pooled plasma and to vWF from plasma of patients with hemophilia A. The kinetic parameters in this report allow estimation of the time needed for complex formation in vivo in healthy individuals and in patients with hemophilia A, in which monoclonally purified or recombinant factor VIII associates with endogenous vWF. Using the plasma concentration of vWF (50 nmol/L in monomers) and the obtained values for K(on) and K(off), the time needed to bind 50% of factor VIII is approximately 2 seconds.


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 ◽  
1996 ◽  
Vol 87 (5) ◽  
pp. 1809-1816 ◽  
Author(s):  
AJ Vlot ◽  
SJ Koppelman ◽  
JC Meijers ◽  
C Dama ◽  
HM van den Berg ◽  
...  

Abstract The binding of factor VIII to von Willebrand factor (vWF) is essential for the protection of factor VIII against proteolytic degradation in plasma. We have characterized the binding kinetics of human factor VIII with vWF using a centrifugation binding assay. Purified or plasma vWF was immobilized with a monoclonal antibody (MoAb RU1) covalently linked to Sepharose (Pharmacia LKB Biotechnology, Uppsala, Sweden). Factor VIII was incubated with vWF-RU1-Sepharose and unbound factor VIII was separated from bound factor VIII by centrifugation. The amount of bound factor VIII was determined from the decrease of factor VIII activity in the supernatant. Factor VIII binding to vWF-RU1-Sepharose conformed to the Langmuir model for independent binding sites with a Kd of 0.46 +/- 0.12 nmol/L, and a stoichiometry of 1.3 factor VIII molecules per vWF monomer at saturation, suggesting that each vWF subunit contains a binding site for factor VIII. Competition experiments were performed with a recombinant vWF (deltaA2-rvWF), lacking residues 730 to 910 which contain the epitope for MoAB RU1. DeltaA2-rvWF effectively displaced previously bound factor VIII, confirming that factor VIII binding to vWF-RU1-Sepharose was reversible. To determine the association rate constant (k(on)) and the dissociation rate constant (k(off)), factor VIII was incubated with vWF-RU1-Sepharose for various time intervals. The observed association kinetics conformed to a simple bimolecular association reaction with k(on) = 5.9 +/- 1.9 x 10(6) M(-1) s(-1) and k(off) = 1.6 +/- 1.2 x 10(-3) s(-1) (mean +/- SD). Similar values were obtained from the dissociation kinetics measured after dilution of preformed factor VIII-vWF-RU1-Sepharose complexes. Identical rate constants were obtained for factor VIII binding to vWF from normal pooled plasma and to vWF from plasma of patients with hemophilia A. The kinetic parameters in this report allow estimation of the time needed for complex formation in vivo in healthy individuals and in patients with hemophilia A, in which monoclonally purified or recombinant factor VIII associates with endogenous vWF. Using the plasma concentration of vWF (50 nmol/L in monomers) and the obtained values for K(on) and K(off), the time needed to bind 50% of factor VIII is approximately 2 seconds.


Blood ◽  
2000 ◽  
Vol 96 (3) ◽  
pp. 958-965 ◽  
Author(s):  
Marc Jacquemin ◽  
Renaud Lavend'homme ◽  
Abdellah Benhida ◽  
Beatrijs Vanzieleghem ◽  
Roseline d'Oiron ◽  
...  

Abstract The mechanisms responsible for the low factor VIII (fVIII) activity in the plasma of patients with mild/moderate hemophilia A are poorly understood. In such patients, we have identified a series of fVIII mutations (Ile2098Ser, Ser2119Tyr, Asn2129Ser, Arg2150His, and Pro2153Gln) clustered in the C1 domain and associated with reduced binding of fVIII to von Willebrand factor (vWf). For each patient plasma, the specific activity of mutated fVIII was close to that of normal fVIII. Scatchard analysis showed that the affinity for vWf of recombinant Ile2098Ser, Ser2119Tyr, and Arg2150His fVIII mutants was reduced 8-fold, 80-fold, and 3-fold, respectively, when compared with normal fVIII. Given the importance of vWf for the stability of fVIII in plasma, these findings suggested that the reduction of fVIII binding to vWf resulting from the above-mentioned mutations could contribute to patients' low fVIII plasma levels. We, therefore, analyzed the effect of vWf on fVIII production by Chinese hamster ovary (CHO) cells transfected with expression vectors for recombinant B domain-deleted normal, Ile2098Ser, Ser2119Tyr, and Arg2150His fVIII. These 3 mutations impaired the vWf-dependent accumulation of functional fVIII in culture medium. Analysis of fVIII production by transiently transfected CHO cells indicated that, in addition to the impaired stabilization by vWf, the secretion of functional Ile2098Ser and Arg2150His fVIII was reduced about 2-fold and 6-fold, respectively, by comparison to Ser2119Tyr and normal fVIII. These findings indicate that C1-domain mutations resulting in reduced fVIII binding to vWf are an important cause of mild/moderate hemophilia A.


Blood ◽  
1983 ◽  
Vol 61 (6) ◽  
pp. 1163-1173 ◽  
Author(s):  
JL Moake ◽  
MJ Weinstein ◽  
JH Troll ◽  
LE Chute ◽  
NM Colannino

Abstract The predominant procoagulant factor VIII (VIII:C) form in normal human plasma containing various combinations of anticoagulants and serine/cysteine protease inhibitors is a protein with mol wt 2.6 +/- 0.2 X 10(5). This protein can be detected by 125I-anti-VIII:C Fab binding and gel electrophoresis in the presence and absence of sodium dodecylsulfate (SDS) and is distinct from the subunit of factor VIII/von Willebrand factor (VIII:vWF) multimers. No larger VIII:C form is present in plasma from patients with severe congenital deficiencies of each of the coagulation factors, other than VIII:C. The mol wt approximately 2.6 X 10(5) VIII:C form is, therefore, likely to be the in vivo procoagulant form of VIII:C, rather than a partially proteolyzed, partially activated derivative of a larger precursor. About 60% of this procoagulant mol wt approximately 2.6 X 10(5) VIII:C form in plasma is present in noncovalent complexes with larger VIII:vWF multimers, which attach reversibly to platelet surfaces in the presence of ristocetin. This VIII:vWF-bound protein of mol wt approximately 2.6 X 10(5) may be the plasma procoagulant form of VIII:C which, after proteolytic activation, accelerates the IXa-mediated cleavage and activation of X postulated to occur on platelet surfaces.


2012 ◽  
Vol 101 (6) ◽  
pp. 2055-2065 ◽  
Author(s):  
Dipak S. Pisal ◽  
Matthew P. Kosloski ◽  
C. Russell Middaugh ◽  
Richard B. Bankert ◽  
Sathy V. Balu-iyer

Blood ◽  
1982 ◽  
Vol 59 (6) ◽  
pp. 1272-1278 ◽  
Author(s):  
ZM Ruggeri ◽  
PM Mannucci ◽  
R Lombardi ◽  
AB Federici ◽  
TS Zimmerman

Abstract We have studied the modifications in the multimeric composition of plasma factor VIII/von Willebrand factor and the bleeding time response following administration of 1-Deamino-[8-D-arginine]-Vasopressin (DDAVP) to patients with different subtypes of von Willebrand's disease. In type I, all multimers were present in plasma in the resting state, though they were decreased in concentration. Administration of DDAVP resulted in an increased concentration of these forms as well as the appearance of larger forms than were previously present. There was concomitant correction of the bleeding time. In type IIA, large multimers were absent in the resting state, and although DDAVP induced an average threefold increase in the plasma concentration of factor VIII/von Willebrand factor, the larger multimers did not appear and the bleeding time, although shortened, was not corrected. In contrast, the larger multimers that were also absent from type IIB plasma in the resting state rapidly appeared following DDAVP administration. However, their appearance was transitory and the bleeding time, as in IIA patients, was shortened but not corrected. The characteristic multimeric composition of platelet factor VIII/von Willebrand factor in given subtypes predicted the alteration in plasma factor VIII/von Willebrand factor induced by DDAVP. These studies provide evidence that the different subtypes of von Willebrand's disease represent distinct abnormalities of factor VIII/von Willebrand factor. They also suggest that complete hemostatic correction following DDAVP can be routinely expected only in type I von Willebrand's disease, and only if factor VIII/von Willebrand factor can be raised to normal levels.


2017 ◽  
Vol 92 (6) ◽  
pp. E94-E96 ◽  
Author(s):  
Veerle Labarque ◽  
Vanitha Perinparajah ◽  
Vanessa Bouskill ◽  
Ann Marie Stain ◽  
Cindy Wakefield ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1007-1007
Author(s):  
Shannon Meeks ◽  
John F. Healey ◽  
John (Pete) S. Lollar

Abstract Factor VIII (fVIII) inhibitory antibodies (fVIII inhibitors) are a significant source of morbidity in patients with hemophilia A. Approximately 30% of patients with severe hemophilia A will develop inhibitors. Most inhibitors are directed against either the A2 or C2 domains of fVIII. The repertoire of antibodies to the C2 domain is functionally complex, including antibodies that inhibit fVIII binding to phospholipid and von Willebrand factor and display either type I or type II kinetics. The goal of this study was to investigate the diversity of the immune response to the C2 domain of human fVIII in a murine hemophilia A model and to identify a set of non-overlapping epitopes recognized by these fVIII inhibitors. A panel of 55 murine anti-C2 antibodies was obtained, which included 53 antibodies from anti-fVIII hybridomas produced in our laboratory and previously described antibodies ESH-4 and ESH-8. Nine of the hybridomas were cloned by limiting dilution and the corresponding monoclonal IgG antibodies were purified. IgG from the remaining 44 hybridomas was isolated directly from the hybridoma supernatants and biotinylated. The 9 monoclonal antibodies along with ESH-4 and ESH-8 were used as primary antibodies in a competition ELISA using human fVIII as the antigen. The panel of 55 biotinylated antibodies was used as secondary antibodies. Antibody pairs were classified as having non-overlapping or overlapping epitopes based on whether the binding of the secondary antibody was present or absent, respectively. A basis set of 3 antibodies, 1B5-1B, 3E6-1B, and (2)117-1B, was defined, which consisted of a set of non-overlapping antibodies that as a group competed for binding of human fVIII with all other antibodies. 1B5-1B and 3E6-1B exhibited type I kinetics with Bethesda titers of 950 and 30, respectively. The Bethesda titer of (2)117-1B could not be determined due to its type II behavior. ESH-4 and ESH-8 had the same epitope profiles as 3E6-1B and (2)117-1B, respectively. Of the 52 non-basis set antibodies, 85% overlapped with 1B5-1B, 29% with 3E6-1B, and 56% with (2)117-1B. The majority of the antibodies overlapped with more than one member of the basis set, leading to the identification of five classes of antibodies (see figure). Because of the large number of antibodies characterized, it is unlikely that the frequency of antibodies in any additional classes is high. The elucidation of the structural complexity of the anti-fVIII C2 repertoire should be useful in the characterization of the pathogenicity of C2 inhibitors. Figure Figure


Sign in / Sign up

Export Citation Format

Share Document