Sie sind auf Seite 1von 9

The

n e w e ng l a n d j o u r na l

of

m e dic i n e

original article

Factor VIII Products and Inhibitor


Development in Severe Hemophilia A
Samantha C. Gouw, M.D., Ph.D., Johanna G. van der Bom, M.D., Ph.D.,
Rolf Ljung, M.D., Ph.D., Carmen Escuriola, M.D., Ana R. Cid, M.D.,
Sgolne Claeyssens-Donadel, M.D., Christel van Geet, M.D., Ph.D.,
Gili Kenet, M.D., Anne Mkipernaa, M.D., Ph.D., Angelo Claudio Molinari, M.D.,
Wolfgang Muntean, M.D., Rainer Kobelt, M.D., George Rivard, M.D.,
Elena Santagostino, M.D., Ph.D., Angela Thomas, M.D., Ph.D.,
and H. Marijke van den Berg, M.D., Ph.D.,
for the PedNet and RODIN Study Group*

A bs t r ac t
Background

For previously untreated children with severe hemophilia A, it is unclear whether


the type of factor VIII product administered and switching among products are associated with the development of clinically relevant inhibitory antibodies (inhibitor
development).
Methods

We evaluated 574 consecutive patients with severe hemophilia A (factor VIII activity,
<0.01 IU per milliliter) who were born between 2000 and 2010 and collected data on
all clotting-factor administration for up to 75 exposure days. The primary outcome
was inhibitor development, which was defined as at least two positive inhibitor tests
with decreased in vivo recovery of factor VIII levels.
Results

Inhibitory antibodies developed in 177 of the 574 children (cumulative incidence,


32.4%); 116 patients had a high-titer inhibitory antibody, defined as a peak titer of at
least 5 Bethesda units per milliliter (cumulative incidence, 22.4%). Plasma-derived
products conferred a risk of inhibitor development that was similar to the risk with
recombinant products (adjusted hazard ratio as compared with recombinant products, 0.96; 95% confidence interval [CI], 0.62 to 1.49). As compared with thirdgeneration full-length recombinant products (derived from the full-length complementary DNA sequence of human factor VIII), second-generation full-length products were associated with an increased risk of inhibitor development (adjusted
hazard ratio, 1.60; 95% CI, 1.08 to 2.37). The content of von Willebrand factor in
the products and switching among products were not associated with the risk of
inhibitor development.

The authors affiliations are listed in the


Appendix. Address reprint requests to
Dr. van den Berg at the University Medical Center Utrecht, Julius Center for
Health Sciences and Primary Care, Rm.
No. Stratenum 5.125, P.O. Box 85500,
3508 GA Utrecht, the Netherlands, or at
h.m.vandeberg@umcutrecht.nl.
* Investigators in the European Pediatric
Network for Hemophilia Management
(PedNet) and the Research of Deter
minants of Inhibitor Development
(RODIN) study group are listed in the
Supplementary Appendix, available at
NEJM.org.
N Engl J Med 2013;368:231-9.
DOI: 10.1056/NEJMoa1208024
Copyright 2013 Massachusetts Medical Society.

Conclusions

Recombinant and plasma-derived factor VIII products conferred similar risks of


inhibitor development, and the content of von Willebrand factor in the products
and switching among products were not associated with the risk of inhibitor development. Second-generation full-length recombinant products were associated with
an increased risk, as compared with third-generation products. (Funded by Bayer
Healthcare and Baxter BioScience.)

n engl j med 368;3 nejm.org january 17, 2013

The New England Journal of Medicine


Downloaded from nejm.org on July 5, 2016. For personal use only. No other uses without permission.
Copyright 2013 Massachusetts Medical Society. All rights reserved.

231

The

n e w e ng l a n d j o u r na l

atients with severe hemophilia A


have a deficiency of functional clotting factor VIII (<0.01 IU per milliliter) and have
bleeding in the joints and muscles. To prevent
joint destruction, the current standard of care for
children with severe hemophilia A is primary prophylaxis. This includes regular infusions of factor VIII, which are initiated at the time of the first
episode of bleeding in a joint or earlier, aiming at
the prevention of joint damage.1 However, in about
30% of children, inhibitory antibodies to infused
factor VIII products develop, making usual treatment with factor VIII and prophylaxis impossible.
There are multiple risk factors for the development of inhibitory antibodies (inhibitor development).2-18
It has been suggested that recombinant factor
VIII products are more immunogenic than plasmaderived products. However, the outcomes of numerous studies and systematic reviews have
been contradictory.19-23 The studies have been
limited by the enrollment of small, heterogeneous study populations and the use of several
factor VIII products, and comparisons among
studies have been difficult because of different
study designs.22,24 The inclusion of minimally
treated patients and patients who were still at risk
for subsequent development of inhibitory antibodies has led to an underestimation of the incidence of inhibitor development.25 In addition,
prospective postmarketing studies could not include high-risk children who started bleeding at
an early age, which meant that the risk of inhibitor development was underestimated. Furthermore, small studies with extreme results are
more likely to be published than are those with
less extreme findings.22 For these reasons, three
systematic reviews of the immunogenicity of
factor VIII products resulted in different conclusions.21-23 Randomized trials comparing the immunogenicity of factor VIII products have not yet
been completed.26
A finding that recombinant and plasmaderived products had a differential risk with respect to inhibitor development would influence
both the decision about which type of product to
administer in individual patients and the availability of the preferred product. Therefore,
knowledge of the risk of inhibitor development
associated with recombinant and plasma-derived
products is important for both the individual
patient with hemophilia and the hemophilia
population as a whole. We assessed whether the
232

of

m e dic i n e

type of factor VIII product and switching among


products were associated with inhibitor development in previously untreated children with severe
hemophilia A.

Me thods
Patients

We enrolled consecutive, previously untreated patients with severe hemophilia A (factor VIII activity, <0.01 IU per milliliter) that had been diagnosed in 1 of the 29 participating hemophilia
treatment centers. All the children in the study
were born between January 1, 2000, and January
1, 2010. Children who had been referred to the
centers because of the presence of inhibitory antibodies were excluded from the study. Approval
was obtained from the institutional review board
at each study center. Parents or guardians of all
children provided written informed consent.
Data Collection

We uniformly collected detailed data on all infusions of factor VIII for up to 75 exposure days or
until the development of inhibitory antibodies,
including dates of infusion, doses and brands of
factor VIII products, reasons for treatment, types
of bleeding, extravasation of products, and surgery.
Patients were followed until the development
of a clinically relevant inhibitory antibody or a
cumulative number of 75 exposure days. (After
75 exposure days, inhibitor development becomes
rare [approximately 2 to 5 cases per 1000 patientyears]).27
Outcomes

The primary outcome was the development of


clinically relevant inhibitory antibodies, which
was defined as at least two positive inhibitor titers combined with decreased in vivo recovery of
factor VIII levels up to the 75th exposure day. The
secondary outcome was the development of a
high-titer inhibitor, which was defined as a peak
titer of at least 5 Bethesda units per milliliter up
to the 75th exposure day.28 A positive inhibitor
titer was defined according to the cutoff level of
the inhibitor assay used in the laboratory at each
center. Factor VIII recovery was described as decreased if the level of factor VIII activity was less
than 66% of the expected level 15 minutes after
the infusion of factor VIII. The expected level of
factor VIII activity was calculated according to the
criteria of Lee et al.29

n engl j med 368;3 nejm.org january 17, 2013

The New England Journal of Medicine


Downloaded from nejm.org on July 5, 2016. For personal use only. No other uses without permission.
Copyright 2013 Massachusetts Medical Society. All rights reserved.

Development of Factor VIII Inhibitors in Hemophilia A

In the majority of centers (92%), patients were


routinely screened for inhibitor development after every 1 to 5 exposure days during the first 20
exposure days and at least every 3 months thereafter. At all centers, patients were closely monitored for signs of inhibitor development, and
investigators performed inhibitor and recovery
testing if there was any suspicion that inhibitory
antibodies had developed.
Types of Factor VIII Products

We assessed the incidence of inhibitor development according to the type of product used at
subsequent exposure days (time-varying determinant). We categorized factor VIII products in several ways. First, we compared the inhibitor risk
between plasma-derived factor VIII products and
recombinant products. Second, to investigate
whether the content of von Willebrand factor was
associated with the risk of inhibitor development,
we categorized factor VIII products into products
containing no von Willebrand factor (all recombinant products), products containing less than
0.01 IU of von Willebrand factor antigen per international unit of factor VIII antigen (monoclonal
antibodypurified plasma-derived products), and
products containing 0.01 IU or more of von Willebrand factor per international unit of factor VIII
antigen (other plasma-derived products).30 Third,
we compared inhibitor incidence among the following categories of factor VIII products: plasmaderived products, first-generation full-length recombinant product (derived from the full-length
complementary DNA sequence of human factor
VIII) (Recombinate, Baxter BioScience), secondgeneration B-domaindeleted recombinant product, and second- and third-generation full-length
recombinant products.
We did not evaluate Kogenate (Bayer Healthcare), a first-generation full-length recombinant
product, or Refacto AF (Pfizer), a third-generation
B-domaindeleted product, because of the small
numbers of patients who received these products
(10 patients [7 as first-use product] and 3 patients [3 as first-use product], respectively). The
product type that was used most frequently was
selected as the reference category.
Switching among Products

We evaluated the risk of inhibitor development in


children who were receiving a plasma-derived
product who were then switched to a recombinant product, as compared with those who were

still receiving a plasma-derived product. We similarly assessed the association between switching
among various types of factor VIII products and
the development of inhibitory antibodies.
Study Conduct

The study was supported by unrestricted research


grants from Bayer Healthcare and Baxter Bio
Science. The companies did not have a role in the
study design, data collection, data analysis, or writing of the manuscript. Representatives of the companies reviewed the manuscript before it was submitted for publication. No one who is not an author
contributed to the writing of the manuscript.
Three of the authors (including the first author) designed the study, performed statistical
analyses, interpreted the data, and vouch for the
integrity of the data, the fidelity of the study to
the protocol, and the accuracy of the data analyses.
The first author wrote the first draft of the manuscript. All the other authors collected the data,
critically reviewed the manuscript, and made the
decision to submit the manuscript for publication.
Statistical Analysis

The absolute risk of inhibitor development varies


according to the cumulative number of exposure
days. To account for this varying risk, we used
pooled logistic regression with the cumulative
number of exposure days as the time variable instead of calendar time. We pooled observations
over all exposure days for all patients into a single sample and then used a logistic-regression
model with stratification according to number of
exposure days to relate the risk factors to inhibitor
development. This method accounts for varying
risks according to the cumulative number of exposure days and is equivalent to Cox regression
with exposure days as time-variable and timedependent covariates.31 Relative hazard rates
were interpreted as relative risks.
We calculated both unadjusted and adjusted
hazard ratios, with the latter adjusted for race or
ethnic group; age at first exposure to factor VIII;
reason for first treatment; interval between exposure days; dose of factor VIII; F8 genotype;
and status with respect to family history of hemophilia and inhibitors, history of switching
among product brands, peak treatment episodes
(defined as treatment with factor VIII for bleeding or for surgery on either 3 consecutive days
or 5 consecutive days), a history of major surgery, and regular prophylaxis. Coding details are

n engl j med 368;3 nejm.org january 17, 2013

The New England Journal of Medicine


Downloaded from nejm.org on July 5, 2016. For personal use only. No other uses without permission.
Copyright 2013 Massachusetts Medical Society. All rights reserved.

233

The

n e w e ng l a n d j o u r na l

of

m e dic i n e

efficacy) for previously untreated patients. An


independent statistician who was unaware of
product types repeated all results by means of
Cox proportional-hazards regression models.

648 Patients were eligible

25 Were excluded
5 Did not provide informed consent
10 Did not have available data
8 Were lost to follow-up
1 Died from intracranial hemorrhage
1 Had unknown reason
17 Had pending informed consent

606 Were included

22 Had baseline-only data


6 Were not yet treated
3 Were lost to follow-up
8 Had data of insufficient quality
5 Had unspecified reasons
7 Had no inhibitor development
15 Had unknown inhibitor status

R e sult s
Patients

A total of 648 patients were eligible for the study.


Of these, 17 patients were excluded because of
pending informed consent, and 25 patients were
excluded by the investigators for various other
reasons (Fig. 1). Baseline data were available for
the remaining 606 patients; the analysis included
574 of these patients (94.7%), for whom detailed
exposure data were available. Their characteristics according to the type of factor VIII product
that was first used are presented in Table 1 (see
also Table 1S in the Supplementary Appendix).
Primary Outcome

584 Remained in study

8 Had insufficient data on exposure days


(reached study end point)
2 Had inhibitor development
6 Had no inhibitor development
2 Received unknown factor VIII product
at first exposure

574 Remained in study

Clinically relevant inhibitory antibodies developed


in 177 patients (cumulative incidence, 32.4%; 95%
confidence interval [CI], 28.5 to 36.3). Of these
patients, 116 had high-titer inhibitors (cumulative incidence, 22.4%; 95% CI, 18.8 to 26.0). Inhibitory antibodies developed after a median of
15 exposure days (interquartile range, 10 to 20)
at a median age of 15.5 months (interquartile
range, 10.7 to 19.6).
Plasma-Derived versus Recombinant Products

516 Reached study end point


58 Did not reach study end point
47 Had treatment data updated until
Jan. 9, 2010
11 Did not have treatment data updated
3 Moved to another center
2 Died from intracranial hemorrhage
1 Was lost to follow-up
5 Had unspecified reasons

Plasma-derived products were used on 4018 exposure days, and recombinant products were used
on 25,661 exposure days. Plasma-derived products
carried a risk of inhibitor development that was
similar to the risk with recombinant products (adjusted hazard ratio as compared with recombinant
products, 0.96; 95% CI, 0.62 to 1.49) (Table 2 and
Fig. 2).

Figure 1. Enrollment and Outcomes.

Content of von Willebrand Factor

provided in the Supplementary Appendix, available with the full text of this article at NEJM.org.
To assess whether our findings were robust,
we also compared the incidence of inhibitor development according to the product brands used
at the first exposure to factor VIII (a time-fixed
determinant). We repeated the analyses in the
subgroup of patients who were not included in
registration trials (primary studies of safety and
234

Seven patients received products with a low von


Willebrand factor content on 1 to 11 exposure
days (total, 26 exposure days). Inhibitory antibodies developed in two patients after receiving a
product with a low von Willebrand factor content.
The risk of inhibitor development with products
containing a high amount of von Willebrand factor was similar to the risk with products containing no von Willebrand factor (adjusted hazard
ratio, 0.90; 95% CI, 0.57 to 1.41).

n engl j med 368;3 nejm.org january 17, 2013

The New England Journal of Medicine


Downloaded from nejm.org on July 5, 2016. For personal use only. No other uses without permission.
Copyright 2013 Massachusetts Medical Society. All rights reserved.

n engl j med 368;3 nejm.org january 17, 2013

The New England Journal of Medicine


Downloaded from nejm.org on July 5, 2016. For personal use only. No other uses without permission.
Copyright 2013 Massachusetts Medical Society. All rights reserved.
16 (8.7)
2 (1.1)
6.1 (3.88.5)
167 (91.3)
117 (63.9)
50 (27.3)
16 (8.7)
100 (54.6)
60 (32.8)
10.2 (7.514.1)
125 (68.3)
12.0 (5.022.5)

41 (22.4)
21 (11.5)
33 (18.0)
64 (37.7)
40 (25.2)

4.6 (3.56.5)
137 (87.3)
71 (45.2)
64 (40.8)
22 (14.0)
95 (60.5)
45 (28.7)
9.9 (5.313.5)
110 (70.1)
17.0 (9.825.3)

40 (25.5)
28 (17.8)
46 (29.3)
41 (28.2)
25 (17.9)

Second-Generation
Full-Length
(N=183)

4 (2.5)
0

Third-Generation
Full-Length
(N=157)

17 (29.0)
14 (24.6)

15 (25.4)
9 (15.3)
18 (30.5)

18.0 (11.331.8)

40 (67.8)

35 (59.3)
18 (30.5)
9.7 (3.811.6)

21 (35.6)
11 (18.6)

27 (45.8)

9.3 (8.410.8)
54 (91.5)

25 (42.4)
0

First-Generation
Full-Length
(N=59)

Recombinant Product*

23 (30.3)
13 (18.0)

20 (26.0)
14 (18.2)
28 (36.4)

18.5 (8.026.3)

62 (80.5)

37 (48.1)
20 (26.0)
8.8 (2.914.0)

21 (27.3)
14 (18.2)

42 (54.5)

9.1 (5.710.5)
73 (94.8)

15 (19.5)
1 (1.3)

Second-Generation
B-DomainDeleted
(N=77)

29 (33.1)
21 (25.7)

30 (34.1)
24 (27.3)
16 (18.2)

12 (521)

67 (76.1)

56 (63.6)
28 (31.8)
7.9 (3.712.8)

24 (27.3)
20 (22.7)

44 (50.0)

6.4 (4.29.6)
81 (92.0)

28 (31.8)
17 (19.3)

Plasma-Derived
Product (N=88)

177 (32.4)
116 (22.4)

149 (26.0)
98 (17.1)
144 (25.1)

15 (725)

411 (71.6)

331 (57.7)
172 (30.0)
9.8 (5.413.5)

187 (32.6)
83 (14.5)

304 (53.0)

6.4 (4.08.9)
521 (90.8)

94 (16.4)
20 (3.5)

All Product Types


(N=574)

* Among the recombinant products, data are not included for seven patients who were first treated with Kogenate (Bayer Healthcare), a first-generation full-length product, and three patients
who were first treated with Refacto AF (Wyeth), a third-generation B-domaindeleted product. However, these patients are included in the total number of patients who received any product.
Race was self-reported.
Low-risk genotypes included those with small deletions and insertions, missense mutations, and splice-site mutations. High-risk genotypes included those with large deletions, nonsense mutations, and intron 1 and 22 inversions.
The start of regular prophylaxis was defined as the moment at which at least three consecutive prophylactic infusions of factor VIII had been given within a period of at least 15 days.
A peak treatment episode was defined as treatment with factor VIII for bleeding or for surgery on either at least 3 consecutive days or at least 5 consecutive days.
The values for inhibitor development are cumulative incidences, which were calculated by means of the KaplanMeier method.

Patients who switched product brands no. (%)


Patients who switched from plasma-derived to
recombinant products no. (%)
Median age (interquartile range) yr
White race no. (%)
Family history of hemophilia no. (%)
No
Yes
Negative for inhibitors
Positive for inhibitors
F8 genotype no. (%)
High-risk
Low-risk
Median age at first exposure to factor VIII (interquartile range) mo
Initiation of regular prophylaxis within first 50 exposure days no. (%)
Median cumulative exposure days at start of prophylaxis (interquartile range) no.
History of peak treatment episode on first exposure
day no. (%)
3 days
5 days
History of surgical procedure no. (%)
Inhibitor development no. (%)
Clinically relevant
High-titer

Characteristic

Table 1. Characteristics of the Patients and the Type of Factor VIII Product Administered during the First Treatment.

Development of Factor VIII Inhibitors in Hemophilia A

235

The

of

m e dic i n e

* NA denotes not applicable.


Hazard ratios were adjusted for race or ethnic group; age at first exposure to factor VIII; reason for first treatment; interval between exposure days; dose of factor VIII; F8 genotype;
and status with respect to family history of hemophilia and inhibitors, history of switching among product brands, peak treatment episodes of either at least 3 consecutive days or at
least 5 consecutive days, history of major surgery, and regular prophylaxis.
A first-generation full-length recombinant product, Kogenate (Bayer Healthcare), and a third-generation B-domaindeleted recombinant product, Refacto AF (Wyeth), were used only in
10 and 3 patients on 103 and 163 exposure days, respectively, so the effect of these products on inhibitor development was not studied.
The only first-generation full-length product in this category was Recombinate (Baxter BioScience).

0.92

0.51
1.23 (0.672.28)

0.97 (0.491.91)

1.51 (0.842.71)

0.17

0.93 (0.481.79)

0.56
1.16 (0.701.92)
0.27

4,018

0.97
1.01 (0.601.70)
0.99

1.31 (0.812.11)
4,018

4,491

Plasma-derived

Second-generation B-domaindeleted

1.00 (0.601.65)

4,491

0.82

0.02

0.53
1.26 (0.612.61)

1.79 (1.092.94)
0.12

0.31

1.47 (0.912.38)

1.44 (0.712.90)

9,143

1.12 (0.612.04)

2,464

0.02

0.96

1.60 (1.082.37)

0.99 (0.531.83)

0.11

0.72

1.37 (0.932.01)
9,143

2,464

Second-generation full-length

1.00
NA
1.00
9,297
NA
1.00
NA
1.00
9,297
Third-generation full-length

Recombinant

Specific products

First-generation full-length

NA

0.85

NA
1.00

0.95 (0.561.61)
4,018

NA
1.00

1.24 (0.752.03)

25,661
NA

0.87

1.00

0.96 (0.621.49)

NA

0.54

1.00

4,018

1.14 (0.751.72)

25,661
Recombinant

All recombinant vs. all plasma-derived


products

Plasma-derived

Any Inhibitor Development


Unadjusted
Adjusted
Hazard Ratio P Value Hazard Ratio P Value

0.40

High-Titer Inhibitor Development


No. of
Unadjusted Hazard
Adjusted
Exposure Days
Ratio
P Value Hazard Ratio P Value

Specific Product Types

No. of
Exposure Days
Product

Table 2. Risk of Inhibitor Development, According to the Type of Factor VIII Product.*

236

n e w e ng l a n d j o u r na l

The risk of inhibitor development was similar


among plasma-derived products, first-generation
full-length recombinant products, second-generation B-domaindeleted recombinant products,
and third-generation recombinant products. Firstgeneration recombinant products were associated with an unadjusted hazard ratio of 1.44 (95%
CI, 0.71 to 2.90) for high-titer inhibitor development; however, after adjustment, the hazard ratio was lower. Second-generation full-length recombinant products were associated with a
significantly higher risk of inhibitor development than were third-generation products (adjusted hazard ratio, 1.60; 95% CI, 1.08 to 2.37;
P=0.02); for high-titer inhibitor development,
the adjusted hazard ratio was 1.79 (95% CI, 1.09
to 2.94; P=0.02) (Table 2).
Switching among Products

Details about the analyses of switching among


brands of factor VIII are provided in the Supplementary Appendix. Switching among products
was not associated with the risk of inhibitor development (adjusted hazard ratio as compared
with no switching, 0.99; 95% CI, 0.63 to 1.56)
(Table 3S in the Supplementary Appendix).
Sensitivity Analyses

The results of sensitivity analyses regarding the


use of factor VIII products (plasma-derived vs.
recombinant products and specific product types)
were similar to those of the primary analysis.
Details regarding the sensitivity analyses are provided in the Supplementary Appendix.

Discussion
In this cohort study involving 574 consecutive,
previously untreated children with severe hemophilia A who were born between 2000 and 2010,
recombinant factor VIII products conferred a risk
of inhibitor development that was similar to the
risk conferred by plasma-derived products. The
von Willebrand factor content in factor VIII products was not associated with inhibitor development. Second-generation full-length recombinant
products were associated with a higher risk of
inhibitor development than were third-generation
full-length products. Switching from a plasmaderived product to a recombinant product or
switching among brands of factor VIII products

n engl j med 368;3 nejm.org january 17, 2013

The New England Journal of Medicine


Downloaded from nejm.org on July 5, 2016. For personal use only. No other uses without permission.
Copyright 2013 Massachusetts Medical Society. All rights reserved.

Development of Factor VIII Inhibitors in Hemophilia A

Adjusted Relative Risk (95% CI)

Fu
llLe
ng
1s
io
th
t
n
2n
G
F
d
en
u
llG
e
Le
en
ra
tio
ng
er
at
th
n
io
Fu
n
llBL
D
en
om
gt
h
ai
n
D
ele
Pl
te
as
d
m
aD
er
iv
ed
at

G
en
er

2n
d

3r

G
en
er

at

io

la
s

Al
lP

ec
o

m
bi

na
nt
m
aD
er
iv
ed

Al
lR

did not result in an increased risk of inhibitor


development.
We directly compared the use of recombinant
products and plasma-derived products in one
study cohort. We avoided selection bias by including all consecutive patients who were born
between January 1, 2000, and January 1, 2010.
We excluded all patients who were referred from
nonparticipating hemophilia centers because of
inhibitor development. We used survival analysis
because at the moment of data analysis a number of patients had not yet reached the study end
point and were still at risk for inhibitor development. This enabled us to include all patients up
to the last exposure day and to calculate cumulative incidences. Collection of detailed information on all 75 exposure days allowed us to adjust
the associations for potential confounding factors. These findings were robust in sensitivity
analyses.
Even though we adjusted for potentially confounding factors, we cannot rule out residual
confounding. The observed associations may have
been affected by information bias, if frequencies
and methods of inhibitor screening among centers had differed according to the particular
factor VIII product. However, we would not expect that the cumulative incidence of high-titer
inhibitor development would be influenced by
variations in inhibitor screening because of a lack
of central laboratory testing, since these inhibitory antibodies will always be detected clinically.
Since the associations were similar with respect
to both all clinically relevant inhibitor development and high-titer inhibitor development, information bias would therefore not have played
a major role. Because a relatively small number
of patients were treated with plasma-derived
products and because of the variety of plasmaderived products, we may not have been able to
detect potential differences in the risk of inhibitor development among various plasma-derived
products.
Several reports have suggested that plasmaderived factor VIII products, especially those containing considerable amounts of von Willebrand
factor, are less immunogenic than recombinant
products.19,32,33 However, several systematic reviews have yielded inconclusive results.21-23 Our
results are in agreement with the findings of a
similarly designed study, the Concerted Action on
Neutralizing Antibodies in Severe Hemophilia A

Figure 2. Adjusted Relative Risk of Inhibitor Development, According to


the Type of Factor VIII Product.
Data are for 574 previously untreated children with severe hemophilia A. In
the comparison between all recombinant products and all plasma-derived
products (at left), the reference group was the recombinant products. In
the comparison of specific products (at right), the reference group was
third-generation full-length recombinant products. The only first-generation
full-length recombinant product that was evaluated was Recombinate (Baxter
BioScience). The I bars indicate 95% confidence intervals.

(CANAL) study, in which the risk of inhibitor


development was not clearly lower with plasmaderived products than with recombinant products (relative risk, 0.79; 95% CI, 0.49 to 1.28).20
Unexpectedly, the risk of inhibitor development was 60% higher among children receiving
a second-generation full-length recombinant
product than among those receiving a thirdgeneration full-length product. This association
may be a biased finding (through confounding,
selection bias, or information bias), a chance finding, or a causal effect.
We accounted for bias from confounding by
adjusting the association for multiple potential
confounding factors. We summarized potentially
confounding factors according to the product
type used at the first treatment (Table 1). Children at increased risk for inhibitor development
did not receive second-generation full-length factor VIII products more often than they did thirdgeneration products. Therefore, confounding does
not seem to explain this association.
We avoided selection bias by including all con-

n engl j med 368;3 nejm.org january 17, 2013

The New England Journal of Medicine


Downloaded from nejm.org on July 5, 2016. For personal use only. No other uses without permission.
Copyright 2013 Massachusetts Medical Society. All rights reserved.

237

The

n e w e ng l a n d j o u r na l

secutive patients and by excluding all patients who


were referred to the participating center because
of a known increased risk for inhibitors. In addition, to further confirm the absence of selection
bias, we performed a sensitivity analysis among
patients who were not in a safety and efficacy
trial. Patients who were included in such a trial
might have been at reduced risk for inhibitor development, since they did not have early bleeding.
The observed increase in the risk of inhibitor
development with second-generation full-length
recombinant products as compared with thirdgeneration full-length products is not likely to be
affected by information bias, since it is unlikely
that patients who were treated with a secondgeneration full-length product were more often
screened for inhibitors than those treated with a
third-generation product. Furthermore, we observed a similar association in high-titer development. Thus, selection bias and information bias
do not explain the observed increase in risk in
second-generation full-length recombinant products, as compared with third-generation fulllength products.
This difference in risk between recombinant
products may be due to chance, which seems unlikely, given the precision of our estimate of effect.
But as long as the observation in our study is not
confirmed in other studies, we cannot exclude
the possibility. However, since bias is unlikely
and the probability of a chance finding is low,
the observed increase in the risk of inhibitor
development in patients receiving second-generation full-length factor VIII products may be real.
Other studies including a systematic review23 and reports of the Kogenate Bayer Study

of

m e dic i n e

Group25,34 have not shown significant differences in the risk of inhibitor development
among various recombinant factor VIII products.
In the registration studies, the incidence of inhibitor development may have been underestimated because of the inclusion of patients who
had already been treated with factor VIII on several exposure days and a short follow-up period
for the subgroup of patients who were still at
risk for inhibitor development. There is no
straightforward biologic explanation for a difference in immunogenicity among recombinant
factor VIII products. Further studies are needed
to verify these observations and to identify biologic explanations.
In conclusion, the use of recombinant factor
VIII products in children with severe hemophilia A
did not have a significant effect on the risk of
inhibitor development, as compared with the use
of plasma-derived products, nor was the von
Willebrand factor content of the products or
switching among them associated with the risk
of inhibitor development. An unexpected finding was that second-generation full-length recombinant products were associated with an
increased risk of inhibitor development, as
compared with third-generation products.
Supported by unrestricted research grants from Bayer
Healthcare and Baxter BioScience.
Disclosure forms provided by the authors are available with
the full text of this article at NEJM.org.
We thank the study coordinator, Ella Smink; Emma Smid and
Mojtaba Hashemi for their support in data cleaning; Yves Guillaume, Kate Khair, Karin Lindvall, Monique Spoor, and Bep
Verkerk for their assistance in the study; and J. Michael Soucie
(Division of Blood Disorders, National Center on Birth Defects
and Developmental Disabilities, Centers for Disease Control and
Prevention) for repeating the analyses.

Appendix
The authors affiliations are as follows: the Department of Pediatrics, Wilhelmina Childrens Hospital (S.C.G.), and the Julius Center
for Health Sciences and Primary Care, University Medical Center Utrecht (S.C.G., H.M.B.), Utrecht, and the Department of Clinical
Epidemiology, Leiden University Medical Center, and the Center for Clinical Transfusion Research, Sanquin Foundation, Leiden (J.G.B.)
all in the Netherlands; Lund University, Department of Pediatrics and Malm Center for Thrombosis and Hemostasis, Sknes Universitetssjukhus, Malm, Sweden (R.L.); Department of Pediatrics, J.W. Goethe University Hospital, Frankfurt, Germany (C.E.); Unidad
de Hemostasia y Trombosis, Hospital Universitario y Politcnico La Fe, Valencia, Spain (A.R.C.); Centre Regional dHemophilie, Centre
Hospitalier Universitaire, Toulouse, France (S.C.-D.); the Department of Pediatrics, University Hospitals Leuven, and the Department of
Cardiovascular Sciences, KU Leuven both in Leuven, Belgium (C.G.); National Hemophilia Center, Ministry of Health, Sheba Medical
Center, Tel Hashomer, Israel (G.K.); Hospital for Children and Adolescents, University of Helsinki, Helsinki (A.M.); Dipartimento di
Ematologia ed Oncologia, Unit Trombosi ed Emostasi, Ospedale Pediatrico Giannina Gaslini, Genoa (A.C.M.), and Angelo Bianchi
Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, Milan (E.S.) both in
Italy; Universittsklinik fr Kinder- und Jugendheilkunde, Graz, Austria (W.M.); Hmophiliezentrum, Wabern and Childrens Hospital
of the University of Bern, Bern, Switzerland (R.K.); Division of Hematology/Oncology, Hpital St. Justine, Montreal (G.R.); and Royal
Hospital for Sick Children, Edinburgh (A.T.).

238

n engl j med 368;3 nejm.org january 17, 2013

The New England Journal of Medicine


Downloaded from nejm.org on July 5, 2016. For personal use only. No other uses without permission.
Copyright 2013 Massachusetts Medical Society. All rights reserved.

Development of Factor VIII Inhibitors in Hemophilia A


References
1. Berntorp E, Boulyjenkov V, Brettler D,

et al. Modern treatment of haemophilia.


Bull World Health Organ 1995;73:691-701.
2. Schwaab R, Brackmann HH, Meyer C,
et al. Haemophilia A: mutation type determines risk of inhibitor formation.
Thromb Haemost 1995;74:1402-6.
3. Gouw SC, van den Berg HM, Oldenburg J, et al. F8 gene mutation type and
inhibitor development in patients with severe hemophilia A: systematic review and
meta-analysis. Blood 2012;119:2922-34.
4. Astermark J, Berntorp E, White GC,
Kroner BL. The Malm International
Brother Study (MIBS): further support for
genetic predisposition to inhibitor development in hemophilia patients. Haemophilia 2001;7:267-72.
5. Aledort LM, Dimichele DM. Inhibitors
occur more frequently in African-American
and Latino haemophiliacs. Haemophilia
1998;4:68.
6. Hay CR, Ollier W, Pepper L, et al. HLA
class II profile: a weak determinant of factor VIII inhibitor development in severe
haemophilia A. Thromb Haemost 1997;
77:234-7.
7. Oldenburg J, Picard JK, Schwaab R, et
al. HLA genotype of patients with severe
haemophilia A due to intron 22 inversion
with and without inhibitors of factor VIII.
Thromb Haemost 1997;77:238-42.
8. Astermark J, Oldenburg J, Pavlova A,
Berntorp E, Lefvert AK. Polymorphisms
in the IL10 but not in the IL1beta and IL4
genes are associated with inhibitor development in patients with hemophilia A.
Blood 2006;107:3167-72.
9. Astermark J, Oldenburg J, Carlson J, et
al. Polymorphisms in the TNFA gene and
the risk of inhibitor development in patients with hemophilia A. Blood 2006;108:
3739-45.
10. Astermark J, Wang X, Oldenburg J,
Berntorp E, Lefvert AK. Polymorphisms
in the CTLA-4 gene and inhibitor development in patients with severe hemophilia A.
J Thromb Haemost 2007;5:263-5.
11. Lozier JN, Rosenberg PS, Goedert JJ,
Menashe I. A case-control study reveals
immunoregulatory gene haplotypes that
influence inhibitor risk in severe haemophilia A. Haemophilia 2011;17:641-9.
12. Pavlova A, Delev D, Lacroix-Desmazes
S, et al. Impact of polymorphisms of the
major histocompatibility complex class II,
interleukin-10, tumor necrosis factoralpha and cytotoxic T-lymphocyte antigen-4 genes on inhibitor development in
severe hemophilia A. J Thromb Haemost
2009;7:2006-15.
13. Bafunno V, Santacroce R, Chetta M, et

al. Polymorphisms in genes involved in


autoimmune disease and the risk of FVIII
inhibitor development in Italian patients
with haemophilia A. Haemophilia 2010;
16:469-73.
14. Gouw SC, van der Bom JG, van den
Berg HM. Treatment-related risk factors
of inhibitor development in previously untreated patients with hemophilia A: the
CANAL cohort study. Blood 2007;109:
4648-54.
15. Gouw SC, van den Berg HM, le Cessie
S, van der Bom JG. Treatment characteristics and the risk of inhibitor development:
a multicenter cohort study among previously untreated patients with severe hemophilia A. J Thromb Haemost 2007;5:
1383-90.
16. Kurnik K, Bidlingmaier C, Engl W, et
al. New early prophylaxis regimen that
avoids immunological danger signals can
reduce FVIII inhibitor development. Haemophilia 2010;16:256-62.
17. Maclean PS, Richards M, Williams M,
et al. Treatment related factors and inhibitor development in children with severe haemophilia A. Haemophilia 2011;
17:282-7.
18. Santagostino E, Mancuso ME, Rocino
A, et al. Environmental risk factors for
inhibitor development in children with
haemophilia A: a case-control study. Br J
Haematol 2005;130:422-7.
19. Goudemand
J,
Rothschild
C,
Demiguel V, et al. Influence of the type of
factor VIII concentrate on the incidence of
factor VIII inhibitors in previously untreated patients with severe hemophilia A.
Blood 2006;107:46-51.
20. Gouw SC, van der Bom JG, Auerswald
G, Ettinghausen CE, Tedgrd U, van den
Berg HM. Recombinant versus plasmaderived factor VIII products and the development of inhibitors in previously untreated patients with severe hemophilia A:
the CANAL cohort study. Blood 2007;109:
4693-7.
21. Wight J, Paisley S. The epidemiology
of inhibitors in haemophilia A: a systematic review. Haemophilia 2003;9:418-35.
22. Iorio A, Halimeh S, Holzhauer S, et al.
Rate of inhibitor development in previously untreated hemophilia A patients treated
with plasma-derived or recombinant factor VIII concentrates: a systematic review.
J Thromb Haemost 2010;8:1256-65.
23. Franchini M, Tagliaferri A, Mengoli
C, Cruciani M. Cumulative inhibitor incidence in previously untreated patients
with severe hemophilia A treated with
plasma-derived versus recombinant factor
VIII concentrates: a critical systematic re-

view. Crit Rev Oncol Hematol 2012;81:8293.


24. Scharrer I, Ehrlich HJ. Reported inhibitor incidence in FVIII PUP studies:
comparing apples with oranges? Haemophilia 2004;10:197-8.
25. Kreuz W, Gill JC, Rothschild C, et al.
Full-length sucrose-formulated recombinant factor VIII for treatment of previously
untreated or minimally treated young
children with severe haemophilia A: results of an international clinical investigation. Thromb Haemost 2005;93:457-67.
26. Mannucci PM, Gringeri A, Peyvandi F,
Santagostino E. Factor VIII products and
inhibitor development: the SIPPET study
(Survey of Inhibitors in Plasma-Product
Exposed Toddlers). Haemophilia 2007;13:
Suppl 5:65-8.
27. Hay CR, Palmer B, Chalmers E, et al.
The incidence of factor VIII inhibitors
throughout life in severe hemophilia A in
the United Kingdom. Blood 2011;117:
6367-70.
28. White GC II, Rosendaal F, Aledort
LM, et al. Definitions in hemophilia: recommendation of the scientific subcommittee on factor VIII and factor IX of the
scientific and standardization committee
of the International Society on Thrombosis
and Haemostasis. Thromb Haemost 2001;
85:560.
29. Lee ML, Gomperts ED, Kingdon HS. A
note on the calculation of recovery for
factor VIII infusions. Thromb Haemost
1993;69:87.
30. Vlot AJ, Koppelman SJ, Bouma BN,
Sixma JJ. Factor VIII and von Willebrand
factor. Thromb Haemost 1998;79:456-65.
31. DAgostino RB, Lee ML, Belanger AJ,
Cupples LA, Anderson K, Kannel WB. Relation of pooled logistic regression to time
dependent Cox regression analysis: the
Framingham Heart Study. Stat Med 1990;
9:1501-15.
32. Ettingshausen CE, Kreuz W. Recombinant vs. plasma-derived products, especially those with intact VWF, regarding
inhibitor development. Haemophilia 2006;
12:Suppl 6:102-6.
33. Mancuso ME, Mannucci PM, Rocino A,
Garagiola I, Tagliaferri A, Santagostino
E. Source and purity of factor VIII products as risk factors for inhibitor development in patients with hemophilia A.
J Thromb Haemost 2012;10:781-90.
34. Giangrande PL. Safety and efficacy of
KOGENATE(R) Bayer in previously untreated patients (PUPs) and minimally
treated patients (MTPs). Haemophilia
2002;8:Suppl 2:19-22.
Copyright 2013 Massachusetts Medical Society.

n engl j med 368;3 nejm.org january 17, 2013

The New England Journal of Medicine


Downloaded from nejm.org on July 5, 2016. For personal use only. No other uses without permission.
Copyright 2013 Massachusetts Medical Society. All rights reserved.

239

Das könnte Ihnen auch gefallen