Download as pdf or txt
Download as pdf or txt
You are on page 1of 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.,
Ségolène Claeyssens-Donadel, M.D., Christel van Geet, M.D., Ph.D.,
Gili Kenet, M.D., Anne Mäkipernaa, 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 authors’ affiliations are listed in the
the type of factor VIII product administered and switching among products are as- Appendix. Address reprint requests to
Dr. van den Berg at the University Medi-
sociated with the development of clinically relevant inhibitory antibodies (inhibitor cal Center Utrecht, Julius Center for
development). Health Sciences and Primary Care, Rm.
No. Stratenum 5.125, P.O. Box 85500,
Methods 3508 GA Utrecht, the Netherlands, or at
We evaluated 574 consecutive patients with severe hemophilia A (factor VIII activity, h.m.vandeberg@umcutrecht.nl.
<0.01 IU per milliliter) who were born between 2000 and 2010 and collected data on * Investigators in the European Pediatric
all clotting-factor administration for up to 75 exposure days. The primary outcome Network for Hemophilia Management
was inhibitor development, which was defined as at least two positive inhibitor tests (PedNet) and the Research of Deter­
minants of Inhibitor Development
with decreased in vivo recovery of factor VIII levels. (RODIN) study group are listed in the
Supplementary Appendix, available at
Results
NEJM.org.
Inhibitory antibodies developed in 177 of the 574 children (cumulative incidence,
N Engl J Med 2013;368:231-9.
32.4%); 116 patients had a high-titer inhibitory antibody, defined as a peak titer of at DOI: 10.1056/NEJMoa1208024
least 5 Bethesda units per milliliter (cumulative incidence, 22.4%). Plasma-derived Copyright © 2013 Massachusetts Medical Society.
products conferred a risk of inhibitor development that was similar to the risk with
recombinant products (adjusted hazard ratio as compared with recombinant prod-
ucts, 0.96; 95% confidence interval [CI], 0.62 to 1.49). As compared with third-
generation full-length recombinant products (derived from the full-length comple-
mentary DNA sequence of human factor VIII), second-generation full-length prod-
ucts 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.
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 devel-
opment. 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 231


The New England Journal of Medicine
Downloaded from nejm.org on November 3, 2014. For personal use only. No other uses without permission.
Copyright © 2013 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

P
atients with severe hemophilia A type of factor VIII product and switching among
have a deficiency of functional clotting fac- products were associated with inhibitor develop-
tor VIII (<0.01 IU per milliliter) and have ment in previously untreated children with severe
bleeding in the joints and muscles. To prevent hemophilia A.
joint destruction, the current standard of care for
children with severe hemophilia A is primary pro- Me thods
phylaxis. This includes regular infusions of fac-
tor VIII, which are initiated at the time of the first Patients
episode of bleeding in a joint or earlier, aiming at We enrolled consecutive, previously untreated pa-
the prevention of joint damage.1 However, in about tients with severe hemophilia A (factor VIII activ-
30% of children, inhibitory antibodies to infused ity, <0.01 IU per milliliter) that had been diag-
factor VIII products develop, making usual treat- nosed in 1 of the 29 participating hemophilia
ment with factor VIII and prophylaxis impossible. treatment centers. All the children in the study
There are multiple risk factors for the develop- were born between January 1, 2000, and January
ment of inhibitory antibodies (inhibitor develop- 1, 2010. Children who had been referred to the
ment).2-18 centers because of the presence of inhibitory an-
It has been suggested that recombinant factor tibodies were excluded from the study. Approval
VIII products are more immunogenic than plasma- was obtained from the institutional review board
derived products. However, the outcomes of nu- at each study center. Parents or guardians of all
merous studies and systematic reviews have children provided written informed consent.
been contradictory.19-23 The studies have been
limited by the enrollment of small, heteroge- Data Collection
neous study populations and the use of several We uniformly collected detailed data on all infu-
factor VIII products, and comparisons among sions of factor VIII for up to 75 exposure days or
studies have been difficult because of different until the development of inhibitory antibodies,
study designs.22,24 The inclusion of minimally including dates of infusion, doses and brands of
treated patients and patients who were still at risk factor VIII products, reasons for treatment, types
for subsequent development of inhibitory anti- of bleeding, extravasation of products, and surgery.
bodies has led to an underestimation of the in- Patients were followed until the development
cidence of inhibitor development.25 In addition, of a clinically relevant inhibitory antibody or a
prospective postmarketing studies could not in- cumulative number of 75 exposure days. (After
clude high-risk children who started bleeding at 75 exposure days, inhibitor development becomes
an early age, which meant that the risk of in- rare [approximately 2 to 5 cases per 1000 patient-
hibitor development was underestimated. Fur- years]).27
thermore, small studies with extreme results are
more likely to be published than are those with Outcomes
less extreme findings.22 For these reasons, three The primary outcome was the development of
systematic reviews of the immunogenicity of clinically relevant inhibitory antibodies, which
factor VIII products resulted in different conclu- was defined as at least two positive inhibitor ti-
sions.21-23 Randomized trials comparing the im- ters combined with decreased in vivo recovery of
munogenicity of factor VIII products have not yet factor VIII levels up to the 75th exposure day. The
been completed.26 secondary outcome was the development of a
A finding that recombinant and plasma- high-titer inhibitor, which was defined as a peak
derived products had a differential risk with re- titer of at least 5 Bethesda units per milliliter up
spect to inhibitor development would influence to the 75th exposure day.28 A positive inhibitor
both the decision about which type of product to titer was defined according to the cutoff level of
administer in individual patients and the avail- the inhibitor assay used in the laboratory at each
ability of the preferred product. Therefore, center. Factor VIII recovery was described as de-
knowledge of the risk of inhibitor development creased if the level of factor VIII activity was less
associated with recombinant and plasma-derived than 66% of the expected level 15 minutes after
products is important for both the individual the infusion of factor VIII. The expected level of
patient with hemophilia and the hemophilia factor VIII activity was calculated according to the
population as a whole. We assessed whether the criteria of Lee et al.29

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

The New England Journal of Medicine


Downloaded from nejm.org on November 3, 2014. 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 still receiving a plasma-derived product. We sim-
routinely screened for inhibitor development af- ilarly assessed the association between switching
ter every 1 to 5 exposure days during the first 20 among various types of factor VIII products and
exposure days and at least every 3 months there- the development of inhibitory antibodies.
after. At all centers, patients were closely moni-
tored for signs of inhibitor development, and Study Conduct
investigators performed inhibitor and recovery The study was supported by unrestricted research
testing if there was any suspicion that inhibitory grants from Bayer Healthcare and Baxter Bio­
antibodies had developed. Science. The companies did not have a role in the
study design, data collection, data analysis, or writ-
Types of Factor VIII Products ing of the manuscript. Representatives of the com-
We assessed the incidence of inhibitor develop- panies reviewed the manuscript before it was sub-
ment according to the type of product used at mitted for publication. No one who is not an author
subsequent exposure days (time-varying determi- contributed to the writing of the manuscript.
nant). We categorized factor VIII products in sev- Three of the authors (including the first au-
eral ways. First, we compared the inhibitor risk thor) designed the study, performed statistical
between plasma-derived factor VIII products and analyses, interpreted the data, and vouch for the
recombinant products. Second, to investigate integrity of the data, the fidelity of the study to
whether the content of von Willebrand factor was the protocol, and the accuracy of the data analyses.
associated with the risk of inhibitor development, The first author wrote the first draft of the manu-
we categorized factor VIII products into products script. All the other authors collected the data,
containing no von Willebrand factor (all recom- critically reviewed the manuscript, and made the
binant products), products containing less than decision to submit the manuscript for publication.
0.01 IU of von Willebrand factor antigen per inter-
national unit of factor VIII antigen (monoclonal Statistical Analysis
antibody–purified plasma-derived products), and The absolute risk of inhibitor development varies
products containing 0.01 IU or more of von Will- according to the cumulative number of exposure
ebrand factor per international unit of factor VIII days. To account for this varying risk, we used
antigen (other plasma-derived products).30 Third, pooled logistic regression with the cumulative
we compared inhibitor incidence among the fol- number of exposure days as the time variable in-
lowing categories of factor VIII products: plasma- stead of calendar time. We pooled observations
derived products, first-generation full-length re- over all exposure days for all patients into a sin-
combinant product (derived from the full-length gle sample and then used a logistic-regression
complementary DNA sequence of human factor model with stratification according to number of
VIII) (Recombinate, Baxter BioScience), second- exposure days to relate the risk factors to inhibitor
generation B-domain–deleted recombinant prod- development. This method accounts for varying
uct, and second- and third-generation full-length risks according to the cumulative number of ex-
recombinant products. posure days and is equivalent to Cox regression
We did not evaluate Kogenate (Bayer Health- with exposure days as time-variable and time-
care), a first-generation full-length recombinant dependent covariates.31 Relative hazard rates
product, or Refacto AF (Pfizer), a third-generation were interpreted as relative risks.
B-domain–deleted product, because of the small We calculated both unadjusted and adjusted
numbers of patients who received these products hazard ratios, with the latter adjusted for race or
(10 patients [7 as first-use product] and 3 pa- ethnic group; age at first exposure to factor VIII;
tients [3 as first-use product], respectively). The reason for first treatment; interval between ex-
product type that was used most frequently was posure days; dose of factor VIII; F8 genotype;
selected as the reference category. and status with respect to family history of he-
mophilia and inhibitors, history of switching
Switching among Products among product brands, peak treatment episodes
We evaluated the risk of inhibitor development in (defined as treatment with factor VIII for bleed-
children who were receiving a plasma-derived ing or for surgery on either ≥3 consecutive days
product who were then switched to a recombi- or ≥5 consecutive days), a history of major sur-
nant product, as compared with those who were gery, and regular prophylaxis. Coding details are

n engl j med 368;3  nejm.org  january 17, 2013 233


The New England Journal of Medicine
Downloaded from nejm.org on November 3, 2014. For personal use only. No other uses without permission.
Copyright © 2013 Massachusetts Medical Society. All rights reserved.
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
648 Patients were eligible product types repeated all results by means of
Cox proportional-hazards regression models.
25 Were excluded
5 Did not provide informed consent
10 Did not have available data
R e sult s
8 Were lost to follow-up
1 Died from intracranial hemorrhage Patients
1 Had unknown reason
17 Had pending informed consent 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
606 Were included
excluded by the investigators for various other
reasons (Fig. 1). Baseline data were available for
22 Had baseline-only data the remaining 606 patients; the analysis included
6 Were not yet treated
3 Were lost to follow-up
574 of these patients (94.7%), for whom detailed
8 Had data of insufficient quality exposure data were available. Their characteris-
5 Had unspecified reasons
tics according to the type of factor VIII product
7 Had no inhibitor development that was first used are presented in Table 1 (see
15 Had unknown inhibitor status
also Table 1S in the Supplementary Appendix).

584 Remained in study Primary Outcome


Clinically relevant inhibitory antibodies developed
in 177 patients (cumulative incidence, 32.4%; 95%
8 Had insufficient data on exposure days
(reached study end point) confidence interval [CI], 28.5 to 36.3). Of these
2 Had inhibitor development patients, 116 had high-titer inhibitors (cumula-
6 Had no inhibitor development
2 Received unknown factor VIII product tive incidence, 22.4%; 95% CI, 18.8 to 26.0). In-
at first exposure hibitory antibodies developed after a median of
15 exposure days (interquartile range, 10 to 20)
574 Remained in study 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
Plasma-derived products were used on 4018 expo-
47 Had treatment data updated until sure days, and recombinant products were used
Jan. 9, 2010
11 Did not have treatment data updated
on 25,661 exposure days. Plasma-derived products
3 Moved to another center carried a risk of inhibitor development that was
2 Died from intracranial hemorrhage
1 Was lost to follow-up
similar to the risk with recombinant products (ad-
5 Had unspecified reasons justed 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
Seven patients received products with a low von
provided in the Supplementary Appendix, avail- Willebrand factor content on 1 to 11 exposure
able with the full text of this article at NEJM.org. days (total, 26 exposure days). Inhibitory anti-
To assess whether our findings were robust, bodies developed in two patients after receiving a
we also compared the incidence of inhibitor de- product with a low von Willebrand factor content.
velopment according to the product brands used The risk of inhibitor development with products
at the first exposure to factor VIII (a time-fixed containing a high amount of von Willebrand fac-
determinant). We repeated the analyses in the tor was similar to the risk with products contain-
subgroup of patients who were not included in ing no von Willebrand factor (adjusted hazard
registration trials (primary studies of safety and ratio, 0.90; 95% CI, 0.57 to 1.41).

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

The New England Journal of Medicine


Downloaded from nejm.org on November 3, 2014. For personal use only. No other uses without permission.
Copyright © 2013 Massachusetts Medical Society. All rights reserved.
Table 1. Characteristics of the Patients and the Type of Factor VIII Product Administered during the First Treatment.

Plasma-Derived All Product Types


Characteristic Recombinant Product* Product (N = 88) (N = 574)
Third-Generation Second-Generation First-Generation Second-Generation
Full-Length Full-Length Full-Length B-Domain–Deleted
(N = 157) (N = 183) (N = 59) (N = 77)
Patients who switched product brands — no. (%) 4 (2.5) 16 (8.7) 25 (42.4) 15 (19.5) 28 (31.8) 94 (16.4)
Patients who switched from plasma-derived to 0 2 (1.1) 0 1 (1.3) 17 (19.3) 20 (3.5)
recombinant products — no. (%)
Median age (interquartile range) — yr 4.6 (3.5–6.5) 6.1 (3.8–8.5) 9.3 (8.4–10.8) 9.1 (5.7–10.5) 6.4 (4.2–9.6) 6.4 (4.0–8.9)
White race — no. (%)† 137 (87.3) 167 (91.3) 54 (91.5) 73 (94.8) 81 (92.0) 521 (90.8)
Family history of hemophilia — no. (%)
No 71 (45.2) 117 (63.9) 27 (45.8) 42 (54.5) 44 (50.0) 304 (53.0)
Yes
Negative for inhibitors 64 (40.8) 50 (27.3) 21 (35.6) 21 (27.3) 24 (27.3) 187 (32.6)
Positive for inhibitors 22 (14.0) 16 (8.7) 11 (18.6) 14 (18.2) 20 (22.7) 83 (14.5)
F8 genotype — no. (%)‡
High-risk 95 (60.5) 100 (54.6) 35 (59.3) 37 (48.1) 56 (63.6) 331 (57.7)
Low-risk 45 (28.7) 60 (32.8) 18 (30.5) 20 (26.0) 28 (31.8) 172 (30.0)
Median age at first exposure to factor VIII (interquar- 9.9 (5.3–13.5) 10.2 (7.5–14.1) 9.7 (3.8–11.6) 8.8 (2.9–14.0) 7.9 (3.7–12.8) 9.8 (5.4–13.5)
tile range) — mo
Initiation of regular prophylaxis within first 50 expo- 110 (70.1) 125 (68.3) 40 (67.8) 62 (80.5) 67 (76.1) 411 (71.6)
sure days — no. (%)§
Median cumulative exposure days at start of prophy- 17.0 (9.8–25.3) 12.0 (5.0–22.5) 18.0 (11.3–31.8) 18.5 (8.0–26.3) 12 (5–21) 15 (7–25)

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

The New England Journal of Medicine


laxis (interquartile range) — no.§
History of peak treatment episode on first exposure
day — no. (%)¶
≥3 days 40 (25.5) 41 (22.4) 15 (25.4) 20 (26.0) 30 (34.1) 149 (26.0)
Development of Factor VIII Inhibitors in Hemophilia A

≥5 days 28 (17.8) 21 (11.5) 9 (15.3) 14 (18.2) 24 (27.3) 98 (17.1)

Copyright © 2013 Massachusetts Medical Society. All rights reserved.


History of surgical procedure — no. (%) 46 (29.3) 33 (18.0) 18 (30.5) 28 (36.4) 16 (18.2) 144 (25.1)
Inhibitor development — no. (%)‖
Clinically relevant 41 (28.2) 64 (37.7) 17 (29.0) 23 (30.3) 29 (33.1) 177 (32.4)
High-titer 25 (17.9) 40 (25.2) 14 (24.6) 13 (18.0) 21 (25.7) 116 (22.4)

Downloaded from nejm.org on November 3, 2014. For personal use only. No other uses without permission.
* 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-domain–deleted 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, non-
sense 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 Kaplan–Meier method.

235
The n e w e ng l a n d j o u r na l of m e dic i n e

Specific Product Types

P Value Hazard Ratio† P Value

‡ A first-generation full-length recombinant product, Kogenate (Bayer Healthcare), and a third-generation B-domain–deleted recombinant product, Refacto AF (Wyeth), were used only in
The risk of inhibitor development was similar

0.85

0.02
0.53
0.92
0.51

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
NA
NA

† 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;
among plasma-derived products, first-generation

1.79 (1.09–2.94)
1.26 (0.61–2.61)
0.97 (0.49–1.91)
1.23 (0.67–2.28)
0.95 (0.56–1.61)
full-length recombinant products, second-gener-
Adjusted

1.00 ation B-domain–deleted recombinant products,

1.00
and third-generation recombinant products. First-
High-Titer Inhibitor Development

generation recombinant products were associat-


ed with an unadjusted hazard ratio of 1.44 (95%
CI, 0.71 to 2.90) for high-titer inhibitor develop-
0.40

0.12
0.31
0.82
0.17
NA

NA
ment; however, after adjustment, the hazard ra-
tio was lower. Second-generation full-length re-
Unadjusted Hazard

combinant products were associated with a


1.24 (0.75–2.03)

1.47 (0.91–2.38)
1.44 (0.71–2.90)
0.93 (0.48–1.79)
1.51 (0.84–2.71)
significantly higher risk of inhibitor develop-
Ratio

1.00

1.00

ment than were third-generation products (ad-

10 and 3 patients on 103 and 163 exposure days, respectively, so the effect of these products on inhibitor development was not studied.
justed 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
Exposure Days

to 2.94; P = 0.02) (Table 2).


25,661
4,018

9,297
9,143
2,464
4,491
4,018
No. of

Switching among Products


Details about the analyses of switching among
brands of factor VIII are provided in the Supple-
Hazard Ratio P Value Hazard Ratio† P Value

0.87

0.02
0.96
0.97
0.56
NA

NA

mentary Appendix. Switching among products


was not associated with the risk of inhibitor de-
1.60 (1.08–2.37)
0.99 (0.53–1.83)
1.01 (0.60–1.70)
1.16 (0.70–1.92)
0.96 (0.62–1.49)

velopment (adjusted hazard ratio as compared


§ The only first-generation full-length product in this category was Recombinate (Baxter BioScience).
Adjusted

with no switching, 0.99; 95% CI, 0.63 to 1.56)


1.00

1.00

(Table 3S in the Supplementary Appendix).


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

Sensitivity Analyses
0.54

0.11
0.72
0.99
0.27

The results of sensitivity analyses regarding the


NA

NA

use of factor VIII products (plasma-derived vs.


least 5 consecutive days, history of major surgery, and regular prophylaxis.
1.14 (0.75–1.72)

1.37 (0.93–2.01)
1.12 (0.61–2.04)
1.00 (0.60–1.65)
1.31 (0.81–2.11)

recombinant products and specific product types)


Unadjusted

were similar to those of the primary analysis.


1.00

1.00

Details regarding the sensitivity analyses are pro-


vided in the Supplementary Appendix.
Exposure Days

Discussion
No. of

25,661
4,018

9,297
9,143
2,464
4,491
4,018

In this cohort study involving 574 consecutive,


previously untreated children with severe hemo-
philia A who were born between 2000 and 2010,
Second-generation B-domain–deleted
All recombinant vs. all plasma-derived

recombinant factor VIII products conferred a risk


of inhibitor development that was similar to the
Second-generation full-length
First-generation full-length§
Third-generation full-length

risk conferred by plasma-derived products. The


* NA denotes not applicable.

von Willebrand factor content in factor VIII prod-


ucts was not associated with inhibitor develop-
ment. Second-generation full-length recombinant
Specific products
products

products were associated with a higher risk of


Plasma-derived

Plasma-derived
Recombinant‡
Recombinant

inhibitor development than were third-generation


full-length products. Switching from a plasma-
Product

derived product to a recombinant product or


switching among brands of factor VIII products

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

The New England Journal of Medicine


Downloaded from nejm.org on November 3, 2014. 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

did not result in an increased risk of inhibitor


3
development.

Adjusted Relative Risk (95% CI)


We directly compared the use of recombinant
products and plasma-derived products in one
study cohort. We avoided selection bias by in- 2

cluding all consecutive patients who were born


between January 1, 2000, and January 1, 2010.
We excluded all patients who were referred from 1
nonparticipating hemophilia centers because of
inhibitor development. We used survival analysis
because at the moment of data analysis a num- 0
ber of patients had not yet reached the study end

nt

ed

th

th

ed
te
gt
na

ng

ng
iv

iv
ele
en
point and were still at risk for inhibitor develop-

er

er
bi

Le

Le

D
D

D
m

ll-

ll-

ll-

n–
a-

a-
o

Fu

Fu
ment. This enabled us to include all patients up

m
ec

ai
s

as
lR

om
la

io

io

tio

Pl
Al

lP
to the last exposure day and to calculate cumula-

at

at

D
ra
Al

B-
er

er

e
en

en

en

n
tive incidences. Collection of detailed informa-

io
G

at
d

t
1s

er
3r

2n
tion on all 75 exposure days allowed us to adjust

en
G
the associations for potential confounding fac-

d
2n
tors. These findings were robust in sensitivity
Figure 2. Adjusted Relative Risk of Inhibitor Development, According to
analyses. the Type of Factor VIII Product.
Even though we adjusted for potentially con- Data are for 574 previously untreated children with severe hemophilia A. In
founding factors, we cannot rule out residual the comparison between all recombinant products and all plasma-derived
confounding. The observed associations may have products (at left), the reference group was the recombinant products. In
been affected by information bias, if frequencies the comparison of specific products (at right), the reference group was
and methods of inhibitor screening among cen- third-generation full-length recombinant products. The only first-generation
full-length recombinant product that was evaluated was Recombinate (Baxter
ters had differed according to the particular BioScience). The I bars indicate 95% confidence intervals.
factor VIII product. However, we would not ex-
pect that the cumulative incidence of high-titer
inhibitor development would be influenced by
variations in inhibitor screening because of a lack (CANAL) study, in which the risk of inhibitor
of central laboratory testing, since these inhibi- development was not clearly lower with plasma-
tory antibodies will always be detected clinically. derived products than with recombinant prod-
Since the associations were similar with respect ucts (relative risk, 0.79; 95% CI, 0.49 to 1.28).20
to both all clinically relevant inhibitor develop- Unexpectedly, the risk of inhibitor develop-
ment and high-titer inhibitor development, in- ment was 60% higher among children receiving
formation bias would therefore not have played a second-generation full-length recombinant
a major role. Because a relatively small number product than among those receiving a third-
of patients were treated with plasma-derived generation full-length product. This association
products and because of the variety of plasma- may be a biased finding (through confounding,
derived products, we may not have been able to selection bias, or information bias), a chance find-
detect potential differences in the risk of inhibi- ing, or a causal effect.
tor development among various plasma-derived We accounted for bias from confounding by
products. adjusting the association for multiple potential
Several reports have suggested that plasma- confounding factors. We summarized potentially
derived factor VIII products, especially those con- confounding factors according to the product
taining considerable amounts of von Willebrand type used at the first treatment (Table 1). Chil-
factor, are less immunogenic than recombinant dren at increased risk for inhibitor development
products.19,32,33 However, several systematic re- did not receive second-generation full-length fac-
views have yielded inconclusive results.21-23 Our tor VIII products more often than they did third-
results are in agreement with the findings of a generation products. Therefore, confounding does
similarly designed study, the Concerted Action on not seem to explain this association.
Neutralizing Antibodies in Severe Hemophilia A We avoided selection bias by including all con-

n engl j med 368;3  nejm.org  january 17, 2013 237


The New England Journal of Medicine
Downloaded from nejm.org on November 3, 2014. For personal use only. No other uses without permission.
Copyright © 2013 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

secutive patients and by excluding all patients who Group25,34 — have not shown significant differ-
were referred to the participating center because ences in the risk of inhibitor development
of a known increased risk for inhibitors. In addi- among various recombinant factor VIII products.
tion, to further confirm the absence of selection In the registration studies, the incidence of in-
bias, we performed a sensitivity analysis among hibitor development may have been underesti-
patients who were not in a safety and efficacy mated because of the inclusion of patients who
trial. Patients who were included in such a trial had already been treated with factor VIII on sev-
might have been at reduced risk for inhibitor de- eral exposure days and a short follow-up period
velopment, since they did not have early bleeding. for the subgroup of patients who were still at
The observed increase in the risk of inhibitor risk for inhibitor development. There is no
development with second-generation full-length straightforward biologic explanation for a dif-
recombinant products as compared with third- ference in immunogenicity among recombinant
generation full-length products is not likely to be factor VIII products. Further studies are needed
affected by information bias, since it is unlikely to verify these observations and to identify bio-
that patients who were treated with a second- logic explanations.
generation full-length product were more often In conclusion, the use of recombinant factor
screened for inhibitors than those treated with a VIII products in children with severe hemophilia A
third-generation product. Furthermore, we ob- did not have a significant effect on the risk of
served a similar association in high-titer develop- inhibitor development, as compared with the use
ment. Thus, selection bias and information bias of plasma-derived products, nor was the von
do not explain the observed increase in risk in Willebrand factor content of the products or
second-generation full-length recombinant prod- switching among them associated with the risk
ucts, as compared with third-generation full- of inhibitor development. An unexpected find-
length products. ing was that second-generation full-length re-
This difference in risk between recombinant combinant products were associated with an
products may be due to chance, which seems un- increased risk of inhibitor development, as
likely, given the precision of our estimate of effect. compared with third-generation products.
But as long as the observation in our study is not
Supported by unrestricted research grants from Bayer
confirmed in other studies, we cannot exclude Healthcare and Baxter BioScience.
the possibility. However, since bias is unlikely Disclosure forms provided by the authors are available with
and the probability of a chance finding is low, the full text of this article at NEJM.org.
We thank the study coordinator, Ella Smink; Emma Smid and
the observed increase in the risk of inhibitor Mojtaba Hashemi for their support in data cleaning; Yves Guil-
development in patients receiving second-genera- laume, Kate Khair, Karin Lindvall, Monique Spoor, and Bep
tion full-length factor VIII products may be real. Verkerk for their assistance in the study; and J. Michael Soucie
(Division of Blood Disorders, National Center on Birth Defects
Other studies — including a systematic re- and Developmental Disabilities, Centers for Disease Control and
view23 and reports of the Kogenate Bayer Study Prevention) for repeating the analyses.

Appendix
The authors’ affiliations are as follows: the Department of Pediatrics, Wilhelmina Children’s 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 Malmö Center for Thrombosis and Hemostasis, Skånes Uni-
versitetssjukhus, Malmö, Sweden (R.L.); Department of Pediatrics, J.W. Goethe University Hospital, Frankfurt, Germany (C.E.); Unidad
de Hemostasia y Trombosis, Hospital Universitario y Politécnico La Fe, Valencia, Spain (A.R.C.); Centre Regional d’Hemophilie, 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, Unità 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; Universitätsklinik für Kinder- und Jugendheilkunde, Graz, Austria (W.M.); Hämophiliezentrum, Wabern and Children’s Hospital
of the University of Bern, Bern, Switzerland (R.K.); Division of Hematology/Oncology, Hôpital 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 November 3, 2014. 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, al. Polymorphisms in genes involved in view. Crit Rev Oncol Hematol 2012;81:82-
et al. Modern treatment of haemophilia. autoimmune disease and the risk of FVIII 93.
Bull World Health Organ 1995;73:691-701. inhibitor development in Italian patients 24. Scharrer I, Ehrlich HJ. Reported in-
2. Schwaab R, Brackmann HH, Meyer C, with haemophilia A. Haemophilia 2010; hibitor incidence in FVIII PUP studies:
et al. Haemophilia A: mutation type de- 16:469-73. comparing apples with oranges? Haemo-
termines risk of inhibitor formation. 14. Gouw SC, van der Bom JG, van den philia 2004;10:197-8.
Thromb Haemost 1995;74:1402-6. Berg HM. Treatment-related risk factors 25. Kreuz W, Gill JC, Rothschild C, et al.
3. Gouw SC, van den Berg HM, Olden- of inhibitor development in previously un- Full-length sucrose-formulated recombi-
burg J, et al. F8 gene mutation type and treated patients with hemophilia A: the nant factor VIII for treatment of previously
inhibitor development in patients with se- CANAL cohort study. Blood 2007;109: untreated or minimally treated young
vere hemophilia A: systematic review and 4648-54. children with severe haemophilia A: re-
meta-analysis. Blood 2012;119:2922-34. 15. Gouw SC, van den Berg HM, le Cessie sults of an international clinical investi-
4. Astermark J, Berntorp E, White GC, S, van der Bom JG. Treatment characteris- gation. Thromb Haemost 2005;93:457-67.
Kroner BL. The Malmö International tics and the risk of inhibitor development: 26. Mannucci PM, Gringeri A, Peyvandi F,
Brother Study (MIBS): further support for a multicenter cohort study among previ- Santagostino E. Factor VIII products and
genetic predisposition to inhibitor devel- ously untreated patients with severe he- inhibitor development: the SIPPET study
opment in hemophilia patients. Haemo- mophilia A. J Thromb Haemost 2007;5: (Survey of Inhibitors in Plasma-Product
philia 2001;7:267-72. 1383-90. Exposed Toddlers). Haemophilia 2007;13:
5. Aledort LM, Dimichele DM. Inhibitors 16. Kurnik K, Bidlingmaier C, Engl W, et Suppl 5:65-8.
occur more frequently in African-American al. New early prophylaxis regimen that 27. Hay CR, Palmer B, Chalmers E, et al.
and Latino haemophiliacs. Haemophilia avoids immunological danger signals can The incidence of factor VIII inhibitors
1998;4:68. reduce FVIII inhibitor development. Hae- throughout life in severe hemophilia A in
6. Hay CR, Ollier W, Pepper L, et al. HLA mophilia 2010;16:256-62. the United Kingdom. Blood 2011;117:
class II profile: a weak determinant of fac- 17. Maclean PS, Richards M, Williams M, 6367-70.
tor VIII inhibitor development in severe et al. Treatment related factors and in- 28. White GC II, Rosendaal F, Aledort
haemophilia A. Thromb Haemost 1997; hibitor development in children with se- LM, et al. Definitions in hemophilia: rec-
77:234-7. vere haemophilia A. Haemophilia 2011; ommendation of the scientific subcom-
7. Oldenburg J, Picard JK, Schwaab R, et 17:282-7. mittee on factor VIII and factor IX of the
al. HLA genotype of patients with severe 18. Santagostino E, Mancuso ME, Rocino scientific and standardization committee
haemophilia A due to intron 22 inversion A, et al. Environmental risk factors for of the International Society on Thrombosis
with and without inhibitors of factor VIII. inhibitor development in children with and Haemostasis. Thromb Haemost 2001;
Thromb Haemost 1997;77:238-42. haemophilia A: a case-control study. Br J 85:560.
8. Astermark J, Oldenburg J, Pavlova A, Haematol 2005;130:422-7. 29. Lee ML, Gomperts ED, Kingdon HS. A
Berntorp E, Lefvert AK. Polymorphisms 19. Goudemand J, Rothschild C, note on the calculation of recovery for
in the IL10 but not in the IL1beta and IL4 Demiguel V, et al. Influence of the type of factor VIII infusions. Thromb Haemost
genes are associated with inhibitor devel- factor VIII concentrate on the incidence of 1993;69:87.
opment in patients with hemophilia A. factor VIII inhibitors in previously un- 30. Vlot AJ, Koppelman SJ, Bouma BN,
Blood 2006;107:3167-72. treated patients with severe hemophilia A. Sixma JJ. Factor VIII and von Willebrand
9. Astermark J, Oldenburg J, Carlson J, et Blood 2006;107:46-51. factor. Thromb Haemost 1998;79:456-65.
al. Polymorphisms in the TNFA gene and 20. Gouw SC, van der Bom JG, Auerswald 31. D’Agostino RB, Lee ML, Belanger AJ,
the risk of inhibitor development in pa- G, Ettinghausen CE, Tedgård U, van den Cupples LA, Anderson K, Kannel WB. Re-
tients with hemophilia A. Blood 2006;108: Berg HM. Recombinant versus plasma- lation of pooled logistic regression to time
3739-45. derived factor VIII products and the devel- dependent Cox regression analysis: the
10. Astermark J, Wang X, Oldenburg J, opment of inhibitors in previously un- Framingham Heart Study. Stat Med 1990;
Berntorp E, Lefvert AK. Polymorphisms treated patients with severe hemophilia A: 9:1501-15.
in the CTLA-4 gene and inhibitor develop- the CANAL cohort study. Blood 2007;109: 32. Ettingshausen CE, Kreuz W. Recom-
ment in patients with severe hemophilia A. 4693-7. binant vs. plasma-derived products, espe-
J Thromb Haemost 2007;5:263-5. 21. Wight J, Paisley S. The epidemiology cially those with intact VWF, regarding
11. Lozier JN, Rosenberg PS, Goedert JJ, of inhibitors in haemophilia A: a system- inhibitor development. Haemophilia 2006;
Menashe I. A case-control study reveals atic review. Haemophilia 2003;9:418-35. 12:Suppl 6:102-6.
immunoregulatory gene haplotypes that 22. Iorio A, Halimeh S, Holzhauer S, et al. 33. Mancuso ME, Mannucci PM, Rocino A,
influence inhibitor risk in severe haemo- Rate of inhibitor development in previous- Garagiola I, Tagliaferri A, Santagostino
philia A. Haemophilia 2011;17:641-9. ly untreated hemophilia A patients treated E. Source and purity of factor VIII prod-
12. Pavlova A, Delev D, Lacroix-Desmazes with plasma-derived or recombinant fac- ucts as risk factors for inhibitor develop-
S, et al. Impact of polymorphisms of the tor VIII concentrates: a systematic review. ment in patients with hemophilia A.
major histocompatibility complex class II, J Thromb Haemost 2010;8:1256-65. J Thromb Haemost 2012;10:781-90.
interleukin-10, tumor necrosis factor- 23. Franchini M, Tagliaferri A, Mengoli 34. Giangrande PL. Safety and efficacy of
alpha and cytotoxic T-lymphocyte anti- C, Cruciani M. Cumulative inhibitor inci- KOGENATE(R) Bayer in previously un-
gen-4 genes on inhibitor development in dence in previously untreated patients treated patients (PUPs) and minimally
severe hemophilia A. J Thromb Haemost with severe hemophilia A treated with treated patients (MTPs). Haemophilia
2009;7:2006-15. plasma-derived versus recombinant factor 2002;8:Suppl 2:19-22.
13. Bafunno V, Santacroce R, Chetta M, et VIII concentrates: a critical systematic re- Copyright © 2013 Massachusetts Medical Society.

n engl j med 368;3  nejm.org  january 17, 2013 239


The New England Journal of Medicine
Downloaded from nejm.org on November 3, 2014. For personal use only. No other uses without permission.
Copyright © 2013 Massachusetts Medical Society. All rights reserved.

You might also like