The antihuman factor VIII (fVIII) C2 domain immune response in hemophilia A mice consists of antibodies that can be divided into 5 groups of structural epitopes and 2 groups of functional epitopes. Groups A, AB, and B consist of classical C2 antibodies that inhibit the binding of fVIII to phospholipid and von Willebrand factor. Groups BC and C contain nonclassical C2 antibodies that block the activation of fVIII by thrombin or factor Xa. Group BC antibodies are the most common and display high specific inhibitory activity and type II kinetics. The C2 epitope groups recognized by 26 polyclonal human anti-fVIII inhibitor plasmas were identified by a novel competition enzyme-linked immunosorbent assay using group-specific murine monoclonal antibodies. Most of the anti-C2 inhibitor plasmas inhibited the binding of both classical and nonclassical antibodies. These results suggest that nonclassical anti-C2 antibodies contribute significantly to the pathogenicity of fVIII inhibitors.

Approximately 30% of patients with hemophilia A develop detectable antifactor VIII (fVIII) antibodies in response to infusions of fVIII.1-4  The immune response to fVIII currently is the most significant complication in the management of patients with hemophilia A. In addition, autoimmune antibodies to fVIII can develop in nonhemophiliacs, producing acquired hemophilia A, which frequently produces life- or limb-threatening bleeding. Most inhibitory antibodies are directed at either the 40-kDa A2 or the 15-kDa C2 domains of the A1-A2-B-ap-A3-C1-C2 fVIII sequence.5  fVIII inhibitors can either inhibit fVIII completely or incompletely at saturating concentrations, corresponding to type I and type II behavior, respectively.6  Classical anti-C2 antibodies inhibit binding of fVIIIa to negatively charged phospholipid membranes.7-9  The binding of fVIII to phospholipid membranes and to von Willebrand factor (VWF) is mutually exclusive, and antibodies have been shown to block binding to both phospholipid and/or VWF.10-14  In addition, murine anti-C2 monoclonal antibodies (mAbs)15,16  and anti-C2 antibodies in 2 polyclonal patient plasmas16,17  have been identified that interfere with the activation of fVIII by thrombin or factor Xa

We recently characterized the diversity of a large panel of murine anti-C2 mAbs.18  Five groups of structural epitopes were defined based on patterns of overlapping epitopes. Group A, AB, and B antibodies correspond to classical inhibitors that inhibit the binding of fVIII to phospholipid and VWF. Group BC antibodies are the most frequent and are type II inhibitors with inhibitory titers usually greater than 10 000 Bethesda units per mg immunoglobulin G. These antibodies inhibit the activation of fVIII by thrombin and factor Xa in the presence and absence of VWF. ESH8, a well-characterized murine anti-C2 mAb, which blocks the release of VWF from fVIII after thrombin activation, is a group C mAb.16  In this study, we used murine group-specific antihuman C2 mAbs in a competition enzyme-linked immunosorbent assay (ELISA) to determine whether nonclassical group BC and C antibodies are present in human fVIII inhibitor patients.

fVIII inhibitor plasmas from 26 patients with congenital hemophilia A or acquired hemophilia A were obtained either as described previously19,20  from the Emory Comprehensive Hemophilia Center or from George King Bio-Medical (Overland Park, KS). Recombinant full-length human fVIII was a gift from Baxter Biosciences (Duarte, CA). mAbs ESH-4 (group A) and ESH-8 (group C) were purchased from American Diagnostica (Greenwich, CT). mAbs 3E6 (group A), I109 (group AB), 1B5 (group B), 2-77 (group BC), and 2-117 (group C) were isolated as described previously.18  mAbs were biotinylated as previously described.18 

Anti-fVIII ELISAs were performed as a modification of previously described procedures.18  Briefly, ELISA plates were coated with fVIII, preincubated with 3 μg/mL of a nonbiotinylated murine antihuman C2 “blocking” mAb, followed by addition of various concentrations of biotinylated antihuman C2 mAb diluted one-ninth in test inhibitor plasma or control (severe hemophilia A noninhibitor) plasma (Figure 1). The blocking mAbs used were 2-77, 3E6/2-117, and I109 for biotinylated mAbs ESH4, 1B5, and ESH8, respectively. Bound biotinylated mAb was quantitated using alkaline-phosphatase conjugated streptavidin and p-nitrophenyl-phosphate.

Figure 1

Identification of group BC/C anti-C2 antibodies in polyclonal human fVIII inhibitor plasmas. fVIII was coated on an ELISA plate and preincubated with group AB mAb I109 in both the control (severe hemophilia A noninhibitor plasma; A) and patient plasma (B) assays. Biotinylated group C Ab ESH8 was serially diluted into control plasma or inhibitor plasma. In the case of the control (C), both the group AB mAb and the biotinylated ESH8 can bind. If a saturating concentration of antibodies exists in the patient plasma that, like ESH8, bind to group BC or C epitopes, they will compete with biotinylated ESH8 for binding to fVIII (D).

Figure 1

Identification of group BC/C anti-C2 antibodies in polyclonal human fVIII inhibitor plasmas. fVIII was coated on an ELISA plate and preincubated with group AB mAb I109 in both the control (severe hemophilia A noninhibitor plasma; A) and patient plasma (B) assays. Biotinylated group C Ab ESH8 was serially diluted into control plasma or inhibitor plasma. In the case of the control (C), both the group AB mAb and the biotinylated ESH8 can bind. If a saturating concentration of antibodies exists in the patient plasma that, like ESH8, bind to group BC or C epitopes, they will compete with biotinylated ESH8 for binding to fVIII (D).

Close modal

ELISA titration curves of bound biotinylated mAb binding were fitted to the 4-parameter logistic equation. The mAb concentration required to produce an A405 of 0.5 (EC0.5) was calculated by interpolation on the fitted curve. The corresponding mAb titer is defined as EC0.5−1. The normal range of EC0.5 values for the binding of biotinylated mAbs was estimated by performing 8 replicate mAb titrations of the control plasma. EC0.5 values (mean ± SD) for ESH4, 1B5, and ESH8 in control plasma were 285 (± 39) ng/mL, 32.6 (± 4.0) ng/mL, and 23.1 (± 2.3) ng/mL, respectively. The corresponding normal range of control plasma ELISA titers was defined using EC0.5 values within 2 SDs from the mean.

The ability of human fVIII inhibitors to compete with the binding of biotinylated murine antihuman fVIII mAbs from groups A (mAb ESH4), B (mAb 1B5), or C (mAb 2-117) to fVIII was evaluated by ELISA (Figure 2). A second, nonbiotinylated mAb also was added to target the specificity of the test plasmas. The principle of the assay is illustrated in Figure 1 for the identification of antibodies with group BC/C specificity. Competition with ESH4 was carried out by preincubating the fVIII-coated ELISA plate with 2-77, a group BC mAb. Group BC antibodies inhibit the binding of group B, BC, and C antibodies. Thus, inhibition of biotinylated ESH4 binding to fVIII under these conditions is limited to group A or group AB antibodies. Similarly, competition with 1B5 was carried out by preincubating the fVIII-coated ELISA plate with 3E6, a group A mAb, and 2-117, a group C mAb. The group A and C mAbs collectively inhibit the binding of antibodies from groups A, AB, BC, and C. Thus, inhibition of biotinylated 1B5 binding to fVIII is limited to group B antibodies. Finally, competition with ESH8 was carried out by preincubating the fVIII-coated ELISA plate with I109, a group AB mAb, which inhibits the binding of antibodies from groups A and AB. Thus, inhibition of biotinylated ESH8 binding to fVIII is limited to nonclassical group BC or group C anti-C2 antibodies.

Figure 2

ELISA titers for biotinylated mAb in the presence of human inhibitor plasmas. ELISA titers for the binding of biotinylated mAbs ESH4 (A), 1B5 (B), and ESH8 (C) in the presence of test or control (noninhibitor hemophilia A) plasma were determined as described in “Methods.” The normal range for control plasma is shown as the gray band. A test-to-control ELISA titer ratio less than 2 SDs from the mean of the control plasma was considered significant. Plasmas represented by ● were run 2 or 3 times; error bars represent SDs. SDs are not reported for plasmas whose ELISA titers were lower than lowest dilution in the titration curve. ○ represent plasmas that were only run once because of limited amounts of plasma. (D) Venn diagram representing the overall distribution of C2 antibodies in the 26 patient plasmas.

Figure 2

ELISA titers for biotinylated mAb in the presence of human inhibitor plasmas. ELISA titers for the binding of biotinylated mAbs ESH4 (A), 1B5 (B), and ESH8 (C) in the presence of test or control (noninhibitor hemophilia A) plasma were determined as described in “Methods.” The normal range for control plasma is shown as the gray band. A test-to-control ELISA titer ratio less than 2 SDs from the mean of the control plasma was considered significant. Plasmas represented by ● were run 2 or 3 times; error bars represent SDs. SDs are not reported for plasmas whose ELISA titers were lower than lowest dilution in the titration curve. ○ represent plasmas that were only run once because of limited amounts of plasma. (D) Venn diagram representing the overall distribution of C2 antibodies in the 26 patient plasmas.

Close modal

Figure 2C indicates that most inhibitory plasmas with C2 specificity contain nonclassical group BC or C antibodies. Human structural epitopes typically are larger in size than murine structural epitopes.21,22  Antibodies with these larger footprints could theoretically block both binding to VWF and/or phospholipid and the activation of fVIII. Given the preincubation step in this protocol, it is possible that antibodies with large footprints, which overlapped both the preincubation mAb and the biotinylated mAb, did not bind to fVIII and therefore could not inhibit the binding of the biotinylated mAb. Thus, this method may underestimate the frequency of antibodies with nonclassical behavior.

Murine group BC antibodies inhibit the activation of fVIII and have high specific inhibitory activity in the Bethesda assay,18  in which the inhibitor titer is defined as the dilution of plasma that produces 50% inhibition of fVIII. However, they are type II inhibitors that do not completely inhibit fVIII. Patients with type II inhibitors often have spontaneous, severe bleeding despite having residual fVIII activities that typically are 10% to 30% at saturating inhibitor concentrations. In contrast, fVIII levels as low as 1% provide significant hemostatic benefit in noninhibitor patients. This indicates that the Bethesda assay underestimates the anticoagulant activity of type II inhibitors. The Bethesda assay is based on the one-stage clotting assay, in which the concentration of thrombin produced may be greater than that during in vivo hemostasis. High concentrations of thrombin may overcome the inhibition of fVIII activation by saturating levels of type II inhibitors. Further investigation is needed into the mechanism of action and the in vivo pathogenicity of nonclassical C2 antibodies.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

This work was supported by grants from the National Institutes of Health (HL082609 and HL40921) and Hemophilia of Georgia, Inc (P.L.), National Hemophilia Foundation Clinical Fellowship (S.L.M.), and Hemophilia and Thrombosis Research Society Research Fellowship (S.L.M.)

National Institutes of Health

Contribution: S.L.M. and J.F.H. designed and performed research, analyzed data, and cowrote the paper; E.T.P. and R.T.B. designed and performed research; and P.L. designed research, analyzed data, and cowrote the paper.

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

Correspondence: Pete Lollar, Emory Children's Center, Room 426D, 2015 Uppergate Drive, Atlanta, GA 30322; e-mail: jlollar@emory.edu.

1
Kogenate, Previously Untreated Patient Study Group
Lusher
 
JM
Arkin
 
S
Abildgaard
 
CF
Schwartz
 
RS
Recombinant factor VIII for the treatment of previously untreated patients with hemophilia A: safety, efficacy, and the development of inhibitors.
N Engl J Med
1993
, vol. 
328
 (pg. 
453
-
459
)
2
Bray
 
GL
Gomperts
 
ED
Courtier
 
S
, et al. 
A multicenter study of recombinant factor VIII (Recombinate): safety, efficacy, and inhibitor risk in previously untreated patients with hemophilia A.
Blood
1994
, vol. 
83
 (pg. 
2428
-
2435
)
3
Kreuz
 
W
Ettingshausen
 
CE
Zyschka
 
A
, et al. 
Inhibitor development in previously untreated patients with hemophilia A: a prospective long-term follow-up comparing plasma-derived and recombinant products.
Semin Thromb Hemost
2002
, vol. 
28
 (pg. 
285
-
290
)
4
the Refacto Phase 3 Study Group
Lusher
 
JM
Lee
 
CA
Kessler
 
CM
Bedrosian
 
CL
The safety and efficacy of B-domain deleted recombinant factor VIII concentrate in patients with severe haemophilia A.
Haemophilia
2003
, vol. 
9
 (pg. 
38
-
49
)
5
Prescott
 
R
Nakai
 
H
Saenko
 
EL
, et al. 
The inhibitory antibody response is more complex in hemophilia A patients than in most nonhemophiliacs with fVIII autoantibodies.
Blood
1997
, vol. 
89
 (pg. 
3663
-
3671
)
6
Gawryl
 
MS
Hoyer
 
LW
Inactivation of factor VIII coagulant activity by two different types of human antibodies.
Blood
1982
, vol. 
60
 (pg. 
1103
-
1109
)
7
Arai
 
M
Scandella
 
D
Hoyer
 
LW
Molecular basis of factor-VIII inhibition by human antibodies: antibodies that bind to the factor-VIII light chain prevent the interaction of factor-VIII with phospholipid.
J Clin Invest
1989
, vol. 
83
 (pg. 
1978
-
1984
)
8
Saenko
 
EL
Shima
 
M
Rajalakshmi
 
KJ
Scandella
 
D
A role for the C2 domain of factor binding to von Willebrand factor.
J Biol Chem
1994
, vol. 
269
 (pg. 
11601
-
11605
)
9
Barrow
 
RT
Healey
 
JF
Jacquemin
 
MG
Saint-Remy
 
JM
Lollar
 
P
Antigenicity of putative phospholipid membrane binding residues in factor VIII.
Blood
2001
, vol. 
97
 (pg. 
169
-
174
)
10
Andersson
 
LO
Brown
 
JE
Interaction of factor VIII-von Willebrand factor with phospholipid vesicles.
Biochem J
1981
, vol. 
200
 (pg. 
161
-
167
)
11
Lajmanovich
 
A
Hudry-Clergeon
 
G
Freyssinet
 
JM
Marguerie
 
G
Human Factor VIII procoagulant activity and phospholipid interactions.
Biochim Biophys Acta
1981
, vol. 
678
 (pg. 
132
-
136
)
12
Saenko
 
EL
Scandella
 
D
A mechanism for inhibition of factor VIII binding to phospholipid by von Willebrand factor.
J Biol Chem
1995
, vol. 
270
 (pg. 
13826
-
13833
)
13
Scandella
 
D
Gilbert
 
GE
Shima
 
M
, et al. 
Some human inhibitor antibodies recognize a common epitope corresponding to C2 domain amino acids 2248-2312 which overlap a phospholipid binding site.
Blood
1995
, vol. 
86
 (pg. 
1811
-
1819
)
14
Shima
 
M
Scandella
 
D
Yoshioka
 
A
, et al. 
A factor VIII neutralizing monoclonal antibody and a human inhibitor alloantibody recognizing epitopes in the C2 domain inhibit factor VIII binding to von Willebrand factor and to phosphatidylserine.
Thromb Haemost
1993
, vol. 
69
 (pg. 
240
-
246
)
15
Nogami
 
K
Shima
 
M
Hosokawa
 
K
, et al. 
Role of factor VIII C2 domain in factor VIII binding to factor Xa.
J Biol Chem
1999
, vol. 
274
 (pg. 
31000
-
31007
)
16
Saenko
 
EL
Shima
 
M
Gilbert
 
GE
Scandella
 
D
Slowed release of thrombin-cleaved factor VIII from von Willebrand factor by a monoclonal and human antibody is a novel mechanism for factor VIII inhibition.
J Biol Chem
1996
, vol. 
271
 (pg. 
27424
-
27431
)
17
Nogami
 
K
Shima
 
M
Hosokawa
 
K
, et al. 
Factor VIII C2 domain contains the thrombin-binding site responsible for thrombin-catalyzed cleavage at Arg1689.
J Biol Chem
2000
, vol. 
275
 (pg. 
25774
-
25780
)
18
Meeks
 
SL
Healey
 
JF
Parker
 
ET
Barrow
 
RT
Lollar
 
P
Anti-human factor VIII C2 domain antibodies in hemophilia A mice recognize a functionally complex continuous spectrum of epitopes dominated by inhibitors of factor VIII activation.
Blood
2007
, vol. 
110
 (pg. 
4234
-
4242
)
19
Barrow
 
RT
Healey
 
JF
Gailani
 
D
Scandella
 
D
Lollar
 
P
Reduction of the antigenicity of factor VIII toward complex inhibitory plasmas using multiply-substituted hybrid human/porcine factor VIII molecules.
Blood
2000
, vol. 
95
 (pg. 
557
-
561
)
20
Barrow
 
RT
Lollar
 
P
Neutralization of anti-factor VIII inhibitors by recombinant porcine factor VIII.
J Thromb Haemost
2006
, vol. 
4
 (pg. 
2223
-
2229
)
21
Davies
 
DR
Padlan
 
EA
Antibody-antigen complexes.
Ann Rev Biochem
1990
, vol. 
59
 (pg. 
439
-
473
)
22
Spiegel
 
PC
Jacquemin
 
M
Saint-Remy
 
JM
Stoddard
 
BL
Pratt
 
KP
Structure of the factor VIII C2 domain-immunoglobulin G4κ Fab complex: identification of an inhibitory antibody epitope on the surface of factor VIII.
Blood
2001
, vol. 
98
 (pg. 
13
-
19
)
Sign in via your Institution