Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

464509

2012
TAH412040620712464509Therapeutic Advances in HematologyC Witmer and G Young

Therapeutic Advances in Hematology Review

Factor VIII inhibitors in hemophilia A: Ther Adv Hematol

(2013) 4(1) 59­–72

rationale and latest evidence DOI: 10.1177/


2040620712464509

© The Author(s), 2012.


Reprints and permissions:
Char Witmer and Guy Young http://www.sagepub.co.uk/
journalsPermissions.nav

Abstract:  Factor VIII (FVIII) replacement therapy is the foundation of treatment in hemophilia
A and is effective unless a patient develops an alloantibody (inhibitor) against exogenous FVIII.
Inhibitor development is currently the most significant treatment complication seen in patients
with hemophilia and is associated with considerable morbidity and a decreased quality of
life. The development of an inhibitor is the result of a complex interaction between a patient’s
immune system and genetic and environmental risk factors. The mainstay of treatment is the
eradication of the inhibitor through immune tolerance. This review summarizes the current
evidence regarding inhibitor risk factors, eradication, and hemostatic bypassing agents.

Keywords:  FVIII, FVIII alloantibodies, hemophilia A, immune tolerance, inhibitors, risk factors

Introduction protein [Fulcher et al. 1987; Gilles et al. 1993]. Correspondence to:
Char Witmer, MD, MSCE
Hemophilia A is the most common congenital IgG4 antibodies are predominant and do not Division of Hematology,
severe bleeding disorder and is the result of a defi- fix complement [Fulcher et al. 1987; Gilles et al. The Children’s Hospital of
Philadelphia, 3501 Civic
ciency in the clotting protein factor VIII. Factor 1993; Lollar, 2004]. The formation of a FVIII Center Boulevard, CTRB
VIII (FVIII) deficiency is an X-linked recessive inhibitor is a T-cell dependent event that includes 11th Floor, Room 11026,
Philadelphia, PA 19104,
disorder occurring in 1 in every 5000 male births antigen-presenting cells, B- and T-helper lym- USA
without an ethnic predominance [Soucie et al. phocytes [Astermark, 2006]. Antibodies can be witmer@email.chop.edu
Guy Young, MD
1998]. Currently the mainstay of treatment is the either inhibitory or noninhibitory. FVIII contains Division of Hematology/
replacement of FVIII with the use of either plasma three A domains (A1, A2, A3), one B domain and Oncology, Children’s
Hospital Los Angeles and
or recombinant FVIII concentrates to achieve two C domains (C1, C2). Inhibitory antibodies University of Southern
hemostasis. FVIII replacement is effective unless are primarily directed against the A2, A3 and C2 California Keck School of
Medicine, Los Angeles,
a patient develops an alloantibody (inhibitor) domains [Fulcher et al. 1985; Scandella et al. CA, USA
against the exogenous FVIII. FVIII inhibitors 1989]. Antibody binding at these domains results
interfere with the infused factor concentrates ren- in steric hindrance blocking functional epitopes
dering them ineffective and necessitating the use of FVIII [Saint-Remy et al. 2004]. These func-
of more costly and less effective alternative hemo- tional epitopes include FIX, phospholipid and
static agents [Di Minno et al. 2010; Gringeri et al. von Willebrand factor interaction sites. Antibodies
2003]. Inhibitor development is currently the in inhibitor patients can simultaneously target
most significant treatment complication seen in multiple FVIII epitopes and these epitope targets
patients with hemophilia. While improvements in can change over time [Fulcher et al. 1988].
hemostatic agents for patients with inhibitors have
resulted in decreased mortality, inhibitors are still FVIII inhibitors are classified based on the kinet-
associated with significant morbidity, including a ics and extent of inhibition of FVIII. Type I inhib-
higher rate of bleeding complications, increased itors follow second-order kinetics (dose-dependent
disability and a decreased quality of life[Brown linear inhibition) and completely inactivate FVIII.
et al. 2009; Darby et al. 2004; Di Minno et al. 2010; Type II inhibitors have complex kinetics and
Gringeri et al. 2003; Morfini et al. 2007]. incompletely inactivate FVIII. Type I inhibi-
tors are more common in severe hemophilia.
Type II inhibitors are more common in inhibi-
Definition of an inhibitor tor patients with mild hemophilia or in patients
An inhibitor is a polyclonal high-affinity immuno- without hemophilia who develop an acquired
globulin G (IgG) that is directed against the FVIII FVIII inhibitor.

http://tah.sagepub.com 59
Therapeutic Advances in Hematology 4 (1)

Laboratory characterization of an inhibitor inhibitor is applied if the assay has been greater
The most common methods used to detect and than 5 BU at any time [White et al. 2001].
quantify FVIII inhibitors include the Bethesda
assay or the Nijmegen-modified Bethesda assay
[Kasper et al. 1975; Verbruggen et al. 1995]. Epidemiology
The International Society on Thrombosis and A systematic review regarding the epidemiology
Hemostasis FVIII/FIX subcommittee recom- of inhibitors in hemophilia A reported an overall
mend that the Nijmegen-modified Bethesda inhibitor prevalence of 5–7% and when limited to
assay be used secondary to improved sensitivity patients with severe disease the prevalence is
and specificity [Giles et al. 1998]. These assays much higher at 12–13% [Wight and Paisley,
only detect inhibitors that reduce FVIII activity 2003]. The incidence of new FVIII inhibitors in
(inhibitory). Both assays utilize serial dilutions patients with severe FVIII deficiency is approxi-
of a patient’s plasma that is incubated with equal mately 30% [Lusher et al. 1993]. About 60% of
volumes of normal plasma for 2 h at 37°C [Lee these inhibitors are high titer (>5 BU) and the
et al. 2005]. The residual factor VIII level of the remaining are low titer (<5 BU) [Lusher et al.
incubation mixtures is measured. A positive 1993]. Inhibitors are less common in patients
result is when there is a significant decrease in with mild or moderate hemophilia occurring in
the residual FVIII. The dilutions and residual approximately 3–13% of patients [Addiego et al.
factor VIII are plotted against each other and the 1993; Ehrenforth et al. 1992; Sultan, 1992]. Most
inhibitor titer is obtained by linear regression patients develop an inhibitor within a relatively
[Lee et al. 2005]. By definition one Nijmegen- short time period of exposure with a median of
Bethesda unit reduces the FVIII activity level 9–12 exposure days [Addiego et al. 1993; Bray
by 50%. et al. 1994; Ehrenforth et al. 1992; Lusher et al.
1993]. A recent study from the UK revealed a
There are limitations to laboratory measures of previously unrecognized bimodal peak of inhibi-
inhibitors, including a limited sensitivity for low tor risk in early childhood and old age [Hay et al.
titer inhibitors [<0.4 Nijmegen-Bethesda unit 2011]. Further studies are needed to investigate
(BU)/ml]. There is also a high inter-laboratory this late peak of inhibitor formation and the
variation in the quantitation of inhibitors with a mechanism that underlie this.
high number of false-positive and false-negative
results [Meijer and Verbruggen, 2009]. This is
likely secondary to a variation in how laboratories Clinical presentation
carry out their assays (use a mix of the Bethesda Factor VIII inhibitors can be detected either with
and Nijmegen methods) and a lack of a refer- routine laboratory testing or by clinical presenta-
ence antibody standard [Meijer and Verbruggen, tion. An inhibitor is clinically suspected when a
2009]. These assays are also better at detecting patient experiences bleeding that does not ade-
and measuring type I inhibitors than type II quately respond to hemostatic therapy. It is also
inhibitors. Enzyme-linked immunosorbent or flu- prudent to check an inhibitor titer prior to an
orescent based immune assays can detect both invasive procedure to ensure adequate hemosta-
inhibitory and noninhibitory antibodies and may sis. Currently there is no consensus regarding the
have improved detection for low-titer inhibitors routine surveillance for inhibitors. Many provid-
but further validation is needed to support wide- ers check inhibitor titers at regular intervals
spread use [Dazzi et al. 1996; Ling et al. 2003; within the highest risk period of the first 50 expo-
Zakarija et al. 2011]. sure days and then annually [Kempton and
White, 2009]. There can be drawbacks to routine
Inhibitors are classified into low- or high-responding surveillance, including the detection of transient
inhibitors based on a patient’s peak inhibitor titer inhibitors which can result in significant patient
after repeated FVIII exposure. The International anxiety.
Society on Thrombosis and Hemostasis Scientific
and Standardization committee has recommended
that an inhibitor titer of 5 BU differentiates low- Genetic risk factors for inhibitor formation
from high-responding inhibitors [White et al. 2001].
An antibody titer that is persistently below 5 BU Genetic mutation
despite repeat challenges with factor VIII is consid- A patient’s specific FVIII genetic mutation is the
ered a low-responding inhibitor. A high-responsive most significant risk factor for inhibitor formation.

60 http://tah.sagepub.com
C Witmer and G Young

While the overall incidence of inhibitor formation genotype still demonstrated an increased risk of
in patients with severe hemophilia A is approxi- inhibitor formation in patients of African or
mately 30%, this proportion varies significantly Hispanic descent [Miller et al. 2012]. The mecha-
among the type of factor VIII mutations. Those nisms that account for these racial/ethnic differ-
mutations that are null (large deletions, nonsense ences remain unclear. Viel and colleagues found
mutations and intron 22 inversions) and do not that a difference in FVIII haplotype between
produce a FVIII protein are associated with the patients of African heritage and recombinant
overall highest rates of inhibitor formation (21–88%) FVIII products accounted for this difference [Viel
[Oldenburg et al. 2004]. The intron 22 inversion, et al. 2009]. Critics of the study by Viel and col-
the most common severe FVIII mutation, has an leagues report concerns regarding adequate control
inhibitor incidence of 21% [Oldenburg et al. 2004]. of confounding and how the mutational analysis
Large deletions that involve multiple domains was completed [Eckhardt et al. 2009a; Lacroix-
have the highest proportion of inhibitor forma- Desmazes et al. 2009; Peyvandi et al. 2009; Santos
tion of 88% [Oldenburg et al. 2004]. Mutations et al. 2009; Yang et al. 2009]. Further studies are
that result in a loss of FVIII function but retain required to confirm this finding.
some FVIII production (missense and splice
mutations) have a lower risk of inhibitor forma-
tion (3–10%) [Oldenburg et al. 2004]. Of special Immunologic factors
interest are small deletions or insertions within Individual immune response traits may also affect
stretches of adenine repeats (poly-A run) that a patient’s reaction to exogenous factor VIII.
result in a very low risk of inhibitor formation Explored immunologic factors include the major
(3%) [Oldenburg et al. 2004]. It was discovered histocompatibility complex (MHC) class II sys-
that there is endogenous restoration of the read- tem and polymorphisms of cytokines [interleukins
ing frame that results in a small amount of FVIII (ILs), tumor necrosis factor (TNF)-α]. To date
production that is likely sufficient to induce tol- MHC phenotype has not consistently been dem-
erance [Young et al. 1997]. onstrated as a risk factor for inhibitor formation
[Astermark et al. 2006a; Hay et al. 1997;
A recent meta-analysis of FVIII genetic mutation Oldenburg et al. 1997; Pavlova et al. 2009]. The
and inhibitor development included data from 30 Malmo International Brother Study demonstrated
different studies with a total of 5383 patients with that polymorphisms of the TNF-α gene and IL-10
severe hemophilia A [Gouw et al. 2012]. To date, are associated with an increased risk of inhibitor
this is the largest study on FVIII genetic mutation formation [Astermark et al. 2006a, 2006b].
and inhibitor formation providing more precise Additional studies regarding IL-10 polymor-
estimates of inhibitor risk. Compared with the phisms have revealed inconsistent results [Bafunno
risk of inhibitor formation in inversion 22, et al. 2010; Lozier et al. 2011; Pavlova et al. 2009].
large deletions and nonsense mutations had an Variation in study results is likely secondary to
increased inhibitor risk with an associated odds study design and the specific hemophilia popu-
ratio (OR) of 3.6 [95% confidence interval (CI) lation that is being studied. Further studies are
2.3–5.7]. The risk of inhibitor formation from needed to confirm these findings.
intron 1 inversion (OR 0.9; 95% CI 0.6–1.5) and
splice site mutations (OR 1; 95% CI 0.6–1.5) was
equal to intron 22 inversion. For small deletions/ Treatment-related risk factors for
insertions and missense mutations the risk was inhibitor formation
lower (OR 0.5; 95% CI 0.4–0.6). Inhibitor formation is a complex immune response
that depends not only on the genetic factors dis-
cussed previously but also on treatment-related
Race/ethnicity risk factors. Treatment-related risk factors that
Patients of African or Hispanic heritage have an have been postulated include age at first expo-
increased risk of inhibitor formation [Aledort sure, the intensity of the first exposure, ‘danger
and Dimichele, 1998; Astermark et al. 2001; signals’, prophylaxis, and the type of FVIII product
Carpenter et al. 2012; Ragni et al. 2009a; Scharrer (recombinant versus plasma derived). Interpretation
et al. 1999]. Early studies demonstrating these of the current studies regarding environmental
associations were criticized secondary to a lack risk factors is complicated secondary to retro-
of control for FVIII genotype. A recent study spective designs, variable methodologies and lack
by Miller and colleagues when controlling for of control for confounding factors.

http://tah.sagepub.com 61
Therapeutic Advances in Hematology 4 (1)

There are conflicting data regarding age at first comparing the cumulative incidence from these
treatment as a risk factor for inhibitor formation. studies is the high degree of heterogeneity in patient
Two small cohort studies found an inverse corre- inhibitor risk factors, study methodology, fre-
lation between the age (<6 months) of first expo- quency of inhibitor testing and length of follow up
sure and inhibitor formation but they were not [Mannucci et al. 2012]. Two recent systematic
controlled for risk factors for inhibitor formation reviews attempted to control for this known hetero-
[Lorenzo et al. 2001; van der Bom et al. 2003]. geneity, and were unable to demonstrate a differ-
Additional studies that controlled for known con- ence in inhibitor formation [Franchini et al. 2012;
founders (i.e. FVIII genetic mutation, treatment Iorio et al. 2010]. In the review by Iorio and col-
intensity) were unable to demonstrate an associa- leagues, study design, study period, testing fre-
tion with age [Chalmers et al. 2007; Gouw et al. quency and median follow up explained most of the
2007b; Maclean et al. 2011; Santagostino et al. variability and the source of factor concentrate lost
2005]. It is likely that the association between statistical significance [Iorio et al. 2010]. To address
inhibitor development and age of first factor this very important clinical question a prospective
exposure is explained by early intensive therapy international randomized clinical trial (SIPPET –
[Kempton and White, 2009]. Survey of Inhibitors in Plasma Product Exposed
Toddlers) is currently enrolling patients and is com-
The intensity of the first FVIII exposure is postu- paring inhibitor incidence in previously untreated
lated to be a risk factor for inhibitor formation patients exposed to either plasma or recombinant
because significant cell injury or inflammation factor products [Mannucci et al. 2007].
leads to immunologic ‘danger signals’ that stimu-
late antigen-presenting cells and amplify an immu- The use of early prophylaxis at a young age has
nologic response which could promote inhibitor been proposed as a mechanism to induce toler-
development [Gallucci and Matzinger, 2001; ance through early controlled antigen exposure
Kono and Rock, 2008; Matzinger, 2002]. Several and to minimize the possibility that the first FVIII
studies have demonstrated an increased rate of exposure is in the setting of a ‘danger signal’. Two
inhibitor formation after intensive treatment (i.e. cohort studies, an Italian cohort and the CANAL
surgical procedure or high-frequency treatment) (concerted action on neutralizing antibodies in
[Gouw et al. 2007a, 2007b; Maclean et al. 2011; severe hemophilia A) study, reported a decreased
Ragni et al. 2009a]. However, a case–control study risk of inhibitor formation for patients receiving
by Santagostino and colleagues did not confirm prophylaxis versus on-demand therapy [Gouw
these findings [Santagostino et al. 2005]. Overall it et al. 2007b; Santagostino et al. 2005]. This was
does appear that there is an increased risk of inhib- not confirmed in a cohort study from the UK
itor formation after an intense initial FVIII expo- [Maclean et al. 2011]. A small prospective pilot
sure. This association is not a reason to withhold study in 26 PUPs from Germany were treated
life-saving therapy but should prompt a clinician at with 25 IU/kg/week as soon as bleeding tendency
the time of a significant early exposure to monitor manifested [Kurnik et al. 2010]. The prevalence
closely for the development of an inhibitor. of inhibitor formation was compared with histori-
cal controls with a significant decrease in inhibi-
There is an ongoing debate regarding the immu- tor formation in the study group with only 1/26
nogenicity of factor products with a concern that (3%) developing inhibitors versus 14/30 (46%)
recombinant products have an increased risk of with an associated OR of 0.048 (95% CI 0.001–
inhibitor formation over that of plasma-derived 0.372) [Kurnik et al. 2010].This study, however,
products. This increased immunogenicity is has been criticized for not having contemporane-
hypothesized to be secondary to alterations in ous controls, and the quite spectacular results
posttranslational modifications of FVIII and a lack need to be confirmed in prospective studies.
of von Willebrand binding [Dasgupta et al. 2007;
Grancha et al. 2011; Hironaka et al. 1992; Qadura Table 1 provides a summary of all the known cur-
et al. 2009]. There are conflicting reports in the lit- rent risk factors for inhibitor formation.
erature that have led to this state of uncertainty. In
comparing studies in previously untreated patients
(PUPs) there is a difference in the overall reported Stratification of inhibitor risk
cumulative incidence of inhibitor formation in With the advancement in the understanding of risk
plasma derived (10.3%) versus recombinant factors that underlie inhibitor formation, efforts
(28.7%) [Mannucci et al. 2012]. The difficulty in have been made to create a clinical scoring system

62 http://tah.sagepub.com
C Witmer and G Young

Table 1.  Risk factors for inhibitor formation in hemophilia A.

Genetic risk factors


Genetic mutation: the highest risk is found for null mutations (i.e. large deletions, nonsense mutations and
the intron 22 inversion)
Family history of an inhibitor
African or Hispanic race/ethnicity
Immune response traits: explored immunologic factors include the major histocompatibility complex class
II system and polymorphisms of cytokines (interleukins, tumor necrosis factor α)
Environmental risk factors
Intensity of the first factor VIII exposure: surgical procedure, high frequency treatment
Inconclusive factors further study is needed: - Source of factor VIII: plasma-derived versus recombinant
factor products
                                        - Early prophylaxis: to prevent inhibitor formation

to quantify an individual’s risk. Ter Avest and col- Eradication of inhibitors


leagues created a clinical scoring system that uti- Owing to the significant clinical ramifications of
lizes three clinical factors (family history of inadequate hemostasis in patients with high-titer
inhibitor, gene mutation and intensive treatment FVIII inhibitors, the overarching therapeutic goal is
at initial exposure) to predict inhibitor formation eradication of the inhibitor. At this time the only
[ter Avest et al. 2008]. The risk score is cumula- proven method for eradication is immune tolerance
tive and includes two points for a positive family induction (ITI). ITI is the regular infusion of FVIII
history of inhibitors, two points for a high-risk to induce FVIII antigen-specific tolerance. There is
gene mutation and three points if the initial FVIII currently no consensus regarding the specifics of
treatment is intensive. Patients without any risk ITI treatment, including the factor product source
factors (risk score=0 points) had an inhibitor inci- (plasma derived versus recombinant), factor dose,
dence of 6%, 23% in those with two points and timing or the use of immune modulation.
57% in those patients with more than three points
[ter Avest et al. 2008]. The limitations of this clin- Multiple immune tolerance registries were estab-
ical scoring system include the use of a primarily lished to help determine patient- and treatment-
white cohort and the lack of inclusion of other related factors associated with immune tolerance
well established risk factors. outcome [Coppola et al. 2009; Dimichele, 2009;
Haya et al. 2001; Lenk, 2000; Mariani et al. 1994].
While a risk score can help identify patients at the Overall reported success rates in these registries
highest risk for inhibitor formation we are still left ranged from 51% to 79% [Coppola et al. 2009;
with a quandary as to how to effectively minimize Dimichele, 2009; Haya et al. 2001; Lenk, 2000;
this risk. As mentioned previously, some propose Mariani et al. 1994]. This wide range in success
the institution of early prophylaxis, although fur- rates is likely secondary to a lack of standardiza-
ther data are needed to confirm these findings. tion in study methodologies, treatment protocols,
Others have proposed the use of alternative and eradication definitions. Consistently across
hemostatic products to delay FVIII exposure. these registries a patient’s peak historical FVIII
Rivard and colleagues completed a prospective inhibitor titer (<200 BU) and the inhibitor titer at
trial in 11 pediatric patients with severe hemo- the time of ITI induction (<10 BU) were associ-
philia using recombinant factor VIIa to try and ated with successful immune tolerance. Other pre-
postpone exposure to FVIII until after 2 years dictors have been identified but have not been
of age [Rivard et al. 2005]. Owing to inadequate consistent across studies. Table 2 provides a com-
hemostasis with recombinant factor VIIa, espe- plete list of predictors for ITI.
cially in mouth bleeding, only 3 out of 11 chil-
dren could postpone FVIII exposure until after There are conflicting data regarding the ITI fac-
2 years of age [Rivard et al. 2005], making this tor VIII dose and success of ITI. The International
therapeutic option limited. Challenges remain as Immune Tolerance Registry demonstrated
to the most effective manner in which to mini- improved ITI success with high doses (200 IU/kg)
mize a patient’s risk of inhibitor formation and while the North American and Spanish Immune
further studies are required. Tolerance Registries demonstrated improved

http://tah.sagepub.com 63
Therapeutic Advances in Hematology 4 (1)

Table 2.  Predictors of immune tolerance induction

Predictors of success Predictors of failure


Pre-ITI inhibitor titer <10 BU [Coppola et al. 2009; Interval >5 years between diagnosis of inhibitor
Dimichele, 2009; Haya et al. 2001; Lenk et al. 2000; and start of ITI [Kroner, 1999]
Mariani et al. 1994]
Historical peak inhibitor titer <200 BU [Coppola et al. ITI interruption [Lenk et al. 2000]
2009; Dimichele, 2009; Haya et al. 2001; Lenk et al.
2000; Mariani et al. 1994]
Low factor VIII dose for ITI* [Dimichele, 2009; Low factor VIII dose for ITI* [Mariani et al. 1994]
Haya et al. 2001]
Factor VIII genotype (low-risk mutations: small High peak titer on ITI [Dimichele, 2009; Haya et al.
insertions, deletions or missense) [Coppola et al. 2009] 2001; Lenk et al. 2000; Mariani et al. 1994]

*There are conflicting results regarding a low factor VIII dose for immune tolerance induction and whether it is a predictor
of failure or success.
BU, Bethesda unit; ITI, immune tolerance induction.

success with lower dosing strategies [Dimichele, products and 63–82% in recombinant products
2009; Haya et al. 2001; Mariani et al. 1994]. A [Franchini and Lippi, 2011]. The RESIST (Rescue
meta-analysis of the International and North Immunotolerance Study naive) trial is currently
American Registries found that FVIII dosing enrolling patients and is a randomized clinical trial
did not influence ITI success [Kroner, 1999]. that compares ITI success in high-risk patients
To address this discrepancy the International (age >6 years, peak historical inhibitor >200, pre-
Immune Tolerance Study was completed [Hay ITI titer >10 BU, time between inhibitor forma-
and DiMichele, 2012]. This multicenter rand- tion and ITI >2 years) using von Willebrand factor
omized clinical trial compared high-dose containing FVIII versus recombinant FVIII
(200 IU/kg/day) with low-dose (50 IU/kg three [Gringeri, 2007]. At this time the available evi-
times/week) regimens in patients with severe dence does not support the superiority of one
hemophilia A and high-titer inhibitors (>5 BU). product source over another for use in ITI.
Product choice was left up to the managing clini-
cian; 90% received recombinant FVIII prod- Although not common to most ITI protocols the
ucts. This study was stopped early secondary to use of immune modulation has been reported as a
increased bleeding events in the low-dose arm. method to improve tolerance success. In Sweden
There was no difference in the proportion of the Malmo protocol uses a combination of immu-
ITI success between the two arms but the time noabsorption (to acutely decreased the FVIII
to achieve ITI success was shorter in the high- inhibitor titer), cyclophosphamide, intravenous
dose arm. Because the study was stopped early immunoglobulin and daily high-dose FVIII
it lacked statistical power to demonstrate thera- [Nilsson et al. 1988]. The reported benefit appears
peutic equivalence below the 30% boundary of to be decreased time to tolerance but the overall
equivalence. It appears that a high-dose strategy success rate is comparable to ITI protocols with-
achieves tolerance at a faster rate and this may out the risks associated with immune modulation
explain the associated decreased bleeding rate. [Berntorp et al. 2000]. Recently there has been an
interest in the use of the monoclonal anti-CD20
Controversy also remains regarding the source of antigen (rituximab) that inhibits B cells and inter-
product for immune tolerance and the impact on feres with IgG production. A phase II single-arm
ITI success. The role of von Willebrand factor in clinical trial used rituximab as a single agent in
achieving tolerance is unclear. The current pub- patients with high-titer inhibitors whose condi-
lished data are limited by small sample size, retro- tion had failed to respond to prior ITI attempts.
spective design and lack of control for known The results of this trial were presented as an oral
confounders. A recent comprehensive literature presentation at the 2011 American Society of
review in comparing these studies found no differ- Hematology Annual Meeting [Leissinger et al.
ence in ITI success based on factor product, with 2011b]. The authors reported that only 3 out of
success rates of 75–87% in plasma-derived FVIII, 16 subjects enrolled (18.8%) had a major response
68–91% in von Willebrand factor containing (fall in the inhibitor to <5 BU without an increase

64 http://tah.sagepub.com
C Witmer and G Young

in the inhibitor titer after rechallenge to FVIII) high-responding inhibitors must be treated with
[Leissinger et al. 2011b]. It appears that as a solo bypassing agents. In general, bypassing agents
agent in patients previously treated with inhibi- can achieve hemostasis but they are not as effec-
tors, rituximab had a small effect but further stud- tive as FVIII replacement in patients without an
ies are needed to determine if rituximab could inhibitor. Options for bypass therapy include an
be more effective if used with ITI. activated prothrombin complex concentrate
(aPCC; FEIBA, FVIII inhibitor bypassing activ-
ity; Baxter AG, Vienna, Austria) or recombinant
New strategies for immunomodulation factor VIIa (rFVIIa; NovoSeven, Novo Nordisk
in FVIII inhibitors A/S, Bagsvaerd, Denmark). Porcine factor VIII is
Currently there is ongoing research in mouse not currently available although a recombinant
models focusing on novel products and methods version is in development [Kempton et al. 2012].
to modulate the immune response to factor VIII Both rFVIIa and aPCCs have been demonstrated
[Miao, 2010; Waters and Lillicrap, 2009]. Multiple to be effective in the treatment of bleeding for
methods have been used to manipulate antigen patients with inhibitors [Buchanan and Kevy,
presentation, including oral or nasal administra- 1978; Hilgartner and Knatterud, 1983; Kurczynski
tion of FVIII peptides, infusion of immature den- and Penner, 1974; Lusher et al. 1998; Shapiro
dritic cells or apoptotic fibroblast cells [Qadura et al. 1998; Sjamsoedin et al. 1981]. A randomized
et al. 2008; Ragni et al. 2009b; Rawle et al. 2006; crossover trial of rFVIIa versus FEIBA assessed
Su et al. 2010]. While these methods demonstrated patient reported efficacy 6 h after treatment and
a decrease in the immune response to FVIII they did not demonstrate statistical equivalence
were unable to completely mitigate this response. between the two products. [Astermark et al. 2007].
Diverse strategies have also been explored that However, the confidence interval of the difference
evade the immune response to FVIII. These include only slightly exceeded the 15% boundary (–11.4%
the preparation of less immunogenic FVIII pro- to 15.7%), p = 0.59[Astermark et al. 2007]. The
teins or through the ongoing exposure to FVIII efficacy between products was rated quite dif-
through novel gene therapy methods including ferently by a substantial portion of subjects
neonatal gene transfer, ex vivo transduction of [Astermark et al. 2007.
hematopoietic stem cells or site-specific
FVIII gene expression in platelets [Miao, 2010]. In determining which bypassing product to use in
Additional focus has also been placed on targeted an individual patient it is important to consider
immunosuppression of either the T- or B-cell multiple factors, including the need to avoid
pathways. The most successful of these immuno- FVIII exposure to prevent anamnesis and a
suppressive efforts include those that increase patient’s individual response to therapy. In a
T-regulatory cells using either protein replace- patient with a newly diagnosed inhibitor whom
ment or gene therapy [Miao, 2010]. While much one is waiting for the FVIII inhibitor titer to fall
of this research is currently in mouse models, it is before initiating ITI it is prudent to use rFVIIa
anticipated that many of these protocols warrant because aPCCs contain a small amount of FVIII
further consideration for clinical translational and have been shown to increase the inhibitor
research. At this time it is unclear whether any of titer. It is well recognized that there can be signifi-
these strategies could be used alone, in combina- cant patient variability in their individual hemo-
tion or in concert with current ITI management. static response to bypassing agents. In some
The ultimate goal for immunomodulation ther- patients bleeding can be unresponsive to mono-
apy is a therapy that allows a patient’s immune therapy and may require alternating products
system to function appropriately while mediating [Gringeri et al. 2011a; Schneiderman et al. 2004].
long-term FVIII tolerance. Secondary to the risk of developing thrombosis
or disseminated intravascular coagulation from
alternating bypassing agents, this therapy should
Hemostatic agents for patients with an be used with caution and only in an inpatient
inhibitor setting [Ingerslev and Sorensen, 2011].
Treatment of bleeding in a patient with an inhibi-
tor is based on the classification of the inhibitor. A It is clear that prophylaxis in patients with severe
low-titer and low-responding inhibitor (<5 BU) hemophilia (without an inhibitor) is effective in
can commonly be overcome with higher doses preventing bleeding [Gringeri et al. 2011b; Manco-
of FVIII, while patients with high-titer/ Johnson et al. 2007]. Recently two trials have

http://tah.sagepub.com 65
Therapeutic Advances in Hematology 4 (1)

demonstrated the efficacy of bypassing agents as Risk factors for inhibitor formation in mild hemo-
secondary prophylaxis for patients with inhibitors philia include later age with first factor VIII
[Konkle et al. 2007; Leissinger et al. 2011a]. The exposure, intensity of the factor exposure, family
first trial compared the efficacy of daily rFVIIa history of inhibitors and type of genetic mutation
prophylaxis (90 μg/kg versus 270 μg/kg) in 22 [Eckhardt et al. 2009b; Hay et al. 1998; Kempton
inhibitor patients and demonstrated decreased et al. 2010; Mauser-Bunschoten et al. 2012;
bleeding at both dose levels (45% versus 59%) Sharathkumar et al. 2003]. Multiple studies have
[Konkle et al. 2007]. Subsequent analysis also reported an association between the Arg593Cys
demonstrated an improvement in quality of life genetic mutation and an increased risk of inhibitor
[Hoots et al. 2008]. More recently, a randomized formation [Eckhardt et al. 2009b; Hay et al. 1998;
crossover clinical trial compared the use of an Mauser-Bunschoten et al. 2012]. However, in a
aPCC (85 units/kg on three nonconsecutive days recent study from the USA the Arg593Cys genetic
a week) with on-demand therapy and demon- mutation was not associated with an increased
strated a 62% overall reduction in all bleeding risk of inhibitor formation [Kempton et al. 2010].
episodes [Leissinger et al. 2011a]. It is unclear HLA genotype has also been explored as a risk fac-
which inhibitor patients should be considered for tor for inhibitor formation in patients with mild
prophylaxis and this should be determined on a hemophilia with the Arg593Cys genetic mutation
case by case basis. It is recommended that proph- and no association was found [Bril et al. 2004].
ylaxis be considered for patients whose condition
has failed to respond to ITI and who have recur- There is no established standard of care for the
rent significant bleeding (i.e. a target joint or life- treatment of patients with mild hemophilia and
threatening hemorrhages) [Young et al. 2011]. an inhibitor. Spontaneous resolution has been
The use of bypassing agents for prophylaxis while reported in up to 60% of cases after a median of
a patient is undergoing an initial course of ITI 9 months (range 0.5–46) [Hay et al. 1998].
should be done with caution because a patient’s However, 75% of patients with spontaneous reso-
FVIII level could improve while receiving a lution experienced anamnesis with repeat FVIII
bypassing agent which could be prothrombotic exposure [Hay et al. 1998]. Eradication has also
[Young et al. 2011]. This is the subject of an ongo- been achieved with the use of immune tolerance
ing clinical trial. by itself or combined with immune modulation
(prednisone, cyclophosphamide or rituximab)
[Franchini et al. 2008; Hay et al. 1998; Robbins
Inhibitors in mild hemophilia A et al. 2001; Vlot et al. 2002]. The primary treatment
As discussed previously inhibitor development in strategy for patients with mild hemophilia should
mild hemophilia is less common then severe be inhibitor prevention. A desmopressin (DDAVP)
hemophilia with an estimated incidence of trial should be completed for all patients with mild
3–13% [Addiego et al. 1993; Ehrenforth et al. hemophilia. DDAVP should be used preferentially
1992; Sultan, 1992]. Patients with mild hemo- over factor products in those who are responders.
philia form inhibitors against the exogenous Mutation analysis and family history can also be
infused factor VIII and commonly there is inhibi- used to identify patients at the highest risk.
tor cross reactivity against the patient’s endoge-
nous FVIII decreasing a patient’s baseline FVIII
level. This decrease changes the patient’s bleed- Conclusion
ing phenotype from mild to moderate or severe. In conclusion, the development of an inhibitor is
Inhibitors can be detected through routine the most significant treatment complication in
screening or with a change in a patient’s bleeding hemophilia A and is the result of complex inter-
pattern. Some patients may have bleeding mani- action between a patient’s immune system and
festations that are more commonly seen in genetic and environmental risk factors. Great
patients with an acquired inhibitor, including strides have been made in understanding the
extensive mucocutaneous or urogenital bleed- important risk factors for inhibitor development,
ing. Others may experience bleeding more com- including delineation of risk by genetic mutation,
mon to hemophilia with joint and intramuscular ethnic/racial differences, immunologic factors, and
hemorrhage. Both type I and II inhibitors have intensity of early factor exposure, but further elu-
been reported in mild hemophilia but there cidation is needed. Questions remain regarding
appears to be a predominance of type II inhibi- the product source and the risk of inhibitor for-
tors [Franchini et al. 2010]. mation and we await the results of the SIPPET

66 http://tah.sagepub.com
C Witmer and G Young

trial to help provide further clarification Astermark, J., Donfield, S., DiMichele, D.,
[Mannucci et al. 2007]. Data regarding early Gringeri, A., Gilbert, S., Waters, J. et al. (2007)
prophylaxis and inhibitor prevention are intrigu- A randomized comparison of bypassing agents in
ing but prospective studies are needed to confirm hemophilia complicated by an inhibitor: the FEIBA
NovoSeven Comparative (FENOC) Study. Blood 109:
these findings. ITI is considered standard of care
546–551.
for inhibitor eradication and a recent randomized
clinical trial demonstrated that a high-dose (200 Astermark, J., Oldenburg, J., Carlson, J., Pavlova, A.,
U/kg daily) regimen achieved tolerance faster and Kavakli, K., Berntorp, E. et al. (2006a)
with less bleeding when compared with a low-dose Polymorphisms in the TNFA gene and the risk of
(50 U/kg three times/week) regimen [Hay and inhibitor development in patients with hemophilia A.
Blood 108: 3739–3745.
DiMichele, 2012]. Uncertainty continues about
the ITI product source, timing, dosing regimen, Astermark, J., Oldenburg, J., Pavlova, A., Berntorp, E.
and the use of immune modulation. There is also and Lefvert, A. (2006b) Polymorphisms in the
new evidence to support the use of prophylaxis IL10 but not in the IL1beta and IL4 genes are
with a bypassing agent in patients with inhibitors, associated with inhibitor development in patients with
although it is unclear which inhibitor patients are hemophilia A. Blood 107: 3167–3172.
the best candidates for this therapy [Konkle et al. Bafunno, V., Santacroce, R., Chetta, M., D’Andrea, G.,
2007; Leissinger et al. 2011a; Young et al. 2011]. Pisanelli, D., Sessa, F. et al. (2010) Polymorphisms in
While significant progress has been made in the genes involved in autoimmune disease and the risk of
care of patients with inhibitors there remain many FVIII inhibitor development in Italian patients with
unanswered questions. Ideally in the future we haemophilia A. Haemophilia 16: 469–473.
will be able to accurately predict a patient’s indi- Berntorp, E., Astermark, J. and Carlborg, E. (2000)
vidual risk of inhibitor formation and accordingly Immune tolerance induction and the treatment of
modify their individual treatment plan to mini- hemophilia. Malmo protocol update. Haematologica
mize or abolish their risk of inhibitor formation. 85(10 Suppl.): 48–50; discussion 50-51.
Bray, G., Gomperts, E., Courter, S., Gruppo, R.,
Funding
Gordon, E., Manco-Johnson, M. et al. (1994)
This review article received no specific grant from A multicenter study of recombinant factor VIII
any funding agency in the public, commercial or (recombinate): safety, efficacy, and inhibitor risk in
not-for-profit sectors. previously untreated patients with hemophilia A. The
Recombinate Study Group. Blood 83: 2428–2435.
Conflict of interest statement
Bril, W., MacLean, P., Kaijen, P., van den Brink, E.,
GY has received honoraria as a speaker for Baxter
Lardy, N., Fijnvandraat, K. et al. (2004) HLA
and Novo Nordisk. CW has no conflicts of inter- class II genotype and factor VIII inhibitors in mild
est to declare. haemophilia A patients with an Arg593 to Cys
mutation. Haemophilia 10: 509–514.
Brown, T., Lee, W., Joshi, A. and Pashos, C. (2009)
References Health-related quality of life and productivity impact
Addiego, J., Kasper, C., Abildgaard, C., Hilgartner, M., in haemophilia patients with inhibitors. Haemophilia
Lusher, J., Glader, B. et al. (1993) Frequency of 15: 911–917.
inhibitor development in haemophiliacs treated with Buchanan, G. and Kevy, S. (1978) Use of
low-purity factor VIII. Lancet 342: 462–464. prothrombin complex concentrates in hemophiliacs
Aledort, L. and Dimichele, D. (1998) Inhibitors occur with inhibitors: clinical and laboratory studies.
more frequently in African-American and Latino Pediatrics 62: 767–774.
haemophiliacs. Haemophilia 4: 68. Carpenter, S., Michael Soucie, J., Sterner, S. and
Astermark, J. (2006) Why do inhibitors develop? Presley, R. (2012) Increased prevalence of inhibitors
Principles of and factors influencing the risk for in Hispanic patients with severe haemophilia A
inhibitor development in haemophilia. Haemophilia enrolled in the Universal Data Collection database.
12(Suppl. 3): 52–60. Haemophilia 18: e260–265.
Astermark, J., Berntorp, E., White, G. and Kroner, B. Chalmers, E., Brown, S., Keeling, D., Liesner, R.,
(2001) The Malmo International Brother Study Richards, M., Stirling, D. et al. (2007) Early factor
(MIBS): further support for genetic predisposition VIII exposure and subsequent inhibitor development
to inhibitor development in hemophilia patients. in children with severe haemophilia A. Haemophilia
Haemophilia 7: 267–272. 13: 149–155.

http://tah.sagepub.com 67
Therapeutic Advances in Hematology 4 (1)

Coppola, A., Margaglione, M., Santagostino, E., Franchini, M., Tagliaferri, A., Mengoli, C. and
Rocino, A., Grandone, E., Mannucci, P. et al. (2009) Cruciani, M. (2012) Cumulative inhibitor incidence
Factor VIII gene (F8) mutations as predictors of in previously untreated patients with severe
outcome in immune tolerance induction of hemophilia hemophilia A treated with plasma-derived versus
A patients with high-responding inhibitors. J Thromb recombinant factor VIII concentrates: a critical
Haemost 7: 1809–1815. systematic review. Crit Rev Oncol Hematol 81:
82–93.
Darby, S., Keeling, D., Spooner, R., Wan Kan, S.,
Giangrande, P., Collins, P. et al. (2004) The Fulcher, C., de Graaf Mahoney, S., Roberts, J.,
incidence of factor VIII and factor IX inhibitors in the Kasper, C. and Zimmerman, T. (1985) Localization
hemophilia population of the UK and their effect on of human factor FVIII inhibitor epitopes to two
subsequent mortality, 1977-99. J Thromb Haemost 2: polypeptide fragments. Proc Natl Acad Sci U S A 82:
1047–1054. 7728–7732.
Dasgupta, S., Repesse, Y., Bayry, J., Navarrete, A., Fulcher, C., de Graaf Mahoney, S. and Zimmerman, T.
Wootla, B., Delignat, S. et al. (2007) VWF protects (1987) FVIII inhibitor IgG subclass and
FVIII from endocytosis by dendritic cells and FVIII polypeptide specificity determined by
subsequent presentation to immune effectors. Blood immunoblotting. Blood 69: 1475–1480.
109: 610–612.
Fulcher, C., Lechner, K. and de Graaf Mahoney, S.
Dazzi, F., Tison, T., Vianello, F., Radossi, P., (1988) Immunoblot analysis shows changes in factor
Zerbinati, P., Carraro, P. et al. (1996) High incidence VIII inhibitor chain specificity in factor VIII inhibitor
of anti-FVIII antibodies against non-coagulant patients over time. Blood 72: 1348–1356.
epitopes in haemophilia A patients: a possible role for
Gallucci, S. and Matzinger, P. (2001) Danger signals:
the half-life of transfused FVIII. Br J Haematol 93:
SOS to the immune system. Curr Opin Immunol 13:
688–693.
114–119.
Dimichele, D. (2009) The North American Immune
Tolerance Registry: contributions to the thirty- Giles, A., Verbruggen, B., Rivard, G., Teitel, J.
year experience with immune tolerance therapy. and Walker, I. (1998) A detailed comparison of the
Haemophilia 15: 320–328. performance of the standard versus the Nijmegen
modification of the Bethesda assay in detecting factor
Di Minno, M., Di Minno, G., Di Capua, M., VIII:C inhibitors in the haemophilia A population of
Cerbone, A. and Coppola, A. (2010) Cost of care of Canada. Association of Hemophilia Centre Directors
haemophilia with inhibitors. Haemophilia 16: e190–e201. of Canada. Factor VIII/IX Subcommittee of Scientific
and Standardization Committee of International
Eckhardt, C., Kamphuisen, P. and Fijnvandraat, K.
Society on Thrombosis and Haemostasis. Thromb
(2009a) Inhibitors of factor VIII in hemophilia.
Haemost 79: 872–875.
N Engl J Med 361: 309; author reply 310.
Gilles, J., Arnout, J., Vermylen, J. and Saint-Remy, J.
Eckhardt, C., Menke, L., van Ommen, C., van der
(1993) Anti-factor VIII antibodies of hemophiliac
Lee, J., Geskus, R., Kamphuisen, P. et al. (2009b)
patients are frequently directed towards nonfunctional
Intensive peri-operative use of factor VIII and the
determinants and do not exhibit isotypic restriction.
Arg593-->Cys mutation are risk factors for inhibitor
Blood 82: 2452–2461.
development in mild/moderate hemophilia A.
J Thromb Haemost 7: 930–937. Gouw, S., van den Berg, H., le Cessie, S. and van
Ehrenforth, S., Kreuz, W., Scharrer, I., Linde, R., der Bom, J. (2007a) Treatment characteristics and
Funk, M., Gungor, T. et al. (1992) Incidence of the risk of inhibitor development: a multicenter
development of factor VIII and factor IX inhibitors in cohort study among previously untreated patients
haemophiliacs. Lancet 339: 594–598. with severe hemophilia A. J Thromb Haemost 5:
1383–1390.
Franchini, M., Favaloro, E. and Lippi, G. (2010)
Mild hemophilia A. J Thromb Haemost 8: 421–432. Gouw, S., van den Berg, H., Oldenburg, J.,
Astermark, J., de Groot, P., Margaglione, M. et
Franchini, M. and Lippi, G. (2011) Immune al. (2012) F8 gene mutation type and inhibitor
tolerance induction for patients with severe development in patients with severe hemophilia A:
hemophilia A: a critical literature review. J Thromb systematic review and meta-analysis. Blood 119:
Thrombolysis 32: 439–447. 2922–2934.
Franchini, M., Mengoli, C., Lippi, G., Targher, G., Gouw, S., van der Bom, J. and Marijke van den Berg, H.
Montagnana, M., Salvagno, G. et al. (2008) Immune (2007b) Treatment-related risk factors of inhibitor
tolerance with rituximab in congenital haemophilia development in previously untreated patients with
with inhibitors: a systematic literature review based on hemophilia A: the CANAL cohort study. Blood 109:
individual patients' analysis. Haemophilia 14: 903–912. 4648–4654.

68 http://tah.sagepub.com
C Witmer and G Young

Grancha, S., Navajas, R., Maranon, C., Paradela, A., plasma and from the culture media of recombinant baby
Albar, J. and Jorquera, J. (2011) Incomplete tyrosine hamster kidney cells. J Biol Chem 267: 8012–8020.
1680 sulphation in recombinant FVIII concentrates.
Hoots, W., Ebbesen, L., Konkle, B., Auerswald, G.,
Haemophilia 17: 709–710.
Roberts, H., Weatherall, J. et al. (2008) Secondary
Gringeri, A. (2007) VWF/FVIII concentrates in prophylaxis with recombinant activated factor VII
high-risk immunotolerance: the RESIST study. improves health-related quality of life of haemophilia
Haemophilia 13(Suppl. 5): 73–77. patients with inhibitors. Haemophilia 14: 466–475.
Gringeri, A., Fischer, K., Karafoulidou, A., Ingerslev, J. and Sorensen, B. (2011) Parallel use of
Klamroth, R., Lopez-Fernandez, M. and by-passing agents in haemophilia with inhibitors: a
Mancuso, E. (2011a) Sequential combined bypassing critical review. Br J Haematol 155: 256–262.
therapy is safe and effective in the treatment of
unresponsive bleeding in adults and children with Iorio, A., Halimeh, S., Holzhauer, S., Goldenberg, N.,
haemophilia and inhibitors. Haemophilia 17: 630–635. Marchesini, E., Marcucci, M. et al. (2010) Rate
of inhibitor development in previously untreated
Gringeri, A., Lundin, B., von Mackensen, S., hemophilia A patients treated with plasma-derived or
Mantovani, L. and Mannucci, P. (2011b) A recombinant factor VIII concentrates: a systematic
randomized clinical trial of prophylaxis in children review. J Thromb Haemost 8: 1256–1265.
with hemophilia A (the ESPRIT Study). J Thromb
Haemost 9: 700–710. Kasper, C., Aledort, L., Aronson, D., Counts, R.,
Edson, J., van Eys, J. et al. (1975) Proceedings: A
Gringeri, A., Mantovani, L., Scalone, L. and more uniform measurement of factor VIII inhibitors.
Mannucci, P. (2003) Cost of care and quality of Thromb Diath Haemorrh 34: 612.
life for patients with hemophilia complicated by
inhibitors: the COCIS Study Group. Blood 102: Kempton, C., Abshire, T., Deveras, R., Hoots, W.,
2358–2363. Gill, J., Kessler, C. et al. (2012) Pharmacokinetics
and safety of OBI-1, a recombinant B domain-deleted
Hay, C. and DiMichele, D. (2012) The principal porcine factor VIII, in subjects with haemophilia A.
results of the International Immune Tolerance Study: a Haemophilia 18: 798–804.
randomized dose comparison. Blood 119: 1335–1344.
Kempton, C., Soucie, J., Miller, C., Hooper, C.,
Hay, C., Ludlam, C., Colvin, B., Hill, F., Preston, F., Escobar, M., Cohen, A. et al. (2010) In non-severe
Wasseem, N. et al. (1998) Factor VIII inhibitors hemophilia A the risk of inhibitor after intensive factor
in mild and moderate-severity haemophilia A. UK treatment is greater in older patients: a case-control
Haemophilia Centre Directors Organisation. Thromb study. J Thromb Haemost 8: 2224–2231.
Haemost 79: 762–766.
Kempton, C. and White, G., II, (2009) How we treat
Hay, C., Ollier, W., Pepper, L., Cumming, A., a hemophilia A patient with a factor VIII inhibitor.
Keeney, S., Goodeve, A. et al. (1997) HLA class II Blood 113: 11–17.
profile: a weak determinant of factor VIII inhibitor
development in severe haemophilia A. UKHCDO Konkle, B., Ebbesen, L., Erhardtsen, E., Bianco, R.,
Inhibitor Working Party. Thromb Haemost 77: Lissitchkov, T., Rusen, L. et al. (2007) Randomized,
234–237. prospective clinical trial of recombinant factor VIIa
for secondary prophylaxis in hemophilia patients with
Hay, C., Palmer, B., Chalmers, E., Liesner, R., inhibitors. J Thromb Haemost 5: 1904–1913.
Maclean, R., Rangarajan, S. et al. (2011) Incidence
of factor VIII inhibitors throughout life in severe Kono, H. and Rock, K. (2008) How dying cells alert
hemophilia A in the United Kingdom. Blood 117: the immune system to danger. Nat Rev Immunol 8:
6367–6370. 279–289.

Haya, S., Lopez, M., Aznar, J. and Batlle, J. (2001) Kroner, B. (1999) Comparison of the international
Immune tolerance treatment in haemophilia patients immune tolerance registry and the North American
with inhibitors: the Spanish Registry. Haemophilia 7: immune tolerance registry. Vox Sang 77(Suppl. 1):
154–159. 33–37.
Hilgartner, M. and Knatterud, G. (1983) The Kurczynski, E. and Penner, J. (1974) Activated
use of factor eight inhibitor by-passing activity prothrombin concentrate for patients with factor VIII
(FEIBA immuno) product for treatment of bleeding inhibitors. N Engl J Med 291: 164–167.
episodes in hemophiliacs with inhibitors. Blood 61:
Kurnik, K., Bidlingmaier, C., Engl, W., Chehadeh, H.,
36–40.
Reipert, B. and Auerswald, G. (2010) New early
Hironaka, T., Furukawa, K., Esmon, P., Fournel, M., prophylaxis regimen that avoids immunological
Sawada, S., Kato, M. et al. (1992) Comparative study danger signals can reduce FVIII inhibitor
of the sugar chains of factor VIII purified from human development. Haemophilia 16: 256–262.

http://tah.sagepub.com 69
Therapeutic Advances in Hematology 4 (1)

Lacroix-Desmazes, S., Dimitrov, J. and Repesse, Y. Manco-Johnson, M., Abshire, T., Shapiro, A., Riske, B.,
(2009) Inhibitors of factor VIII in hemophilia. N Engl Hacker, M., Kilcoyne, R. et al. (2007) Prophylaxis
J Med 361: 308; author reply 310. versus episodic treatment to prevent joint disease
in boys with severe hemophilia. N Engl J Med 357:
Lee, C., Berntorp, E. and Hoots, W. (2005) Textbook
535–544.
of Hemophilia. 1st edn. Malden, MA: Blackwell
Publishing. Mannucci, P., Gringeri, A., Peyvandi, F. and
Santagostino, E. (2007) Factor VIII products and
Leissinger, C., Gringeri, A., Antmen, B.,
inhibitor development: the SIPPET study (survey
Berntorp, E., Biasoli, C., Carpenter, S. et al. (2011a)
of inhibitors in plasma-product exposed toddlers).
Anti-inhibitor coagulant complex prophylaxis in
Haemophilia 13(Suppl. 5): 65–68.
hemophilia with inhibitors. N Engl J Med 365:
1684–1692. Mannucci, P., Mancuso, M. and Santagostino,
E. (2012) How we choose factor VIII to treat
Leissinger, C., Kruse-Jarres, R., Granger, S.,
hemophilia. Blood 119: 4108–4114.
Konkle, B., Ragni, M., Journeycake, J. et al. (2011b)
Phase II trial of rituximab in the treatment of Mariani, G., Scheibel, E., Nogao, T., Kasper, C.,
inhibitors in congenital hemophilia A: results of the Ewing, N., Mauser-Bunschoten, E. et al. (1994)
RICH study. Blood (ASH Annual Meeting Abstracts) Immunetolerance as treatment of alloantibodies to
118: 27. factor VIII in hemophilia. The International Registry
of Immunetolerance Protocols. Semin Hematol
Lenk, H. (2000) The German Registry of immune
31(2 Suppl. 4): 62–64.
tolerance treatment in hemophilia – 1999 update.
Haematologica 85(10 Suppl.): 45–47. Matzinger, P. (2002) The danger model: a renewed
sense of self. Science 296: 301–305.
Ling, M., Duncan, E., Rodgers, S., Street, A. and
Lloyd, J. (2003) Low detection rate of antibodies to Mauser-Bunschoten, E., Den Uijl, I., Schutgens, R.,
non-functional epitopes on factor VIII in patients with Roosendaal, G. and Fischer, K. (2012) Risk of
hemophilia A and negative for inhibitors by Bethesda inhibitor development in mild haemophilia A
assay. J Thromb Haemost 1: 2548–2553. increases with age. Haemophilia 18: 263–267.
Lollar, P. (2004) Pathogenic antibodies to coagulation Meijer, P. and Verbruggen, B. (2009) The between-
factors. Part one: factor VIII and factor IX. J Thromb laboratory variation of factor VIII inhibitor testing: the
Haemost 2: 1082–1095. experience of the external quality assessment program
Lorenzo, J., Lopez, A., Altisent, C. and Aznar, J. of the ECAT foundation. Semin Thromb Hemost 35:
(2001) Incidence of factor VIII inhibitors in severe 786–793.
haemophilia: the importance of patient age. Br J Miao, C. (2010) Immunomodulation for inhibitors in
Haematol 113: 600–603. hemophilia A: the important role of Treg cells. Expert
Lozier, J., Rosenberg, P., Goedert, J. and Menashe, I. Rev Hematol 3: 469–483.
(2011) A case-control study reveals immunoregulatory Miller, C., Benson, J., Ellingsen, D., Driggers, J.,
gene haplotypes that influence inhibitor risk in severe Payne, A., Kelly, F. et al. (2012) F8 and F9 mutations
haemophilia A. Haemophilia 17: 641–649. in US haemophilia patients: correlation with history of
Lusher, J., Arkin, S., Abildgaard, C. and Schwartz, R. inhibitor and race/ethnicity. Haemophilia 18: 375–382.
(1993) Recombinant factor VIII for the treatment Morfini, M., Haya, S., Tagariello, G., Pollmann, H.,
of previously untreated patients with hemophilia Quintana, M., Siegmund, B. et al. (2007) European
A. Safety, efficacy, and development of inhibitors. study on orthopaedic status of haemophilia patients
Kogenate Previously Untreated Patient Study Group. with inhibitors. Haemophilia 13: 606–612.
N Engl J Med 328: 453–459.
Nilsson, I., Berntorp, E. and Zettervall, O. (1988)
Lusher, J., Roberts, H., Davignon, G., Joist, J., Induction of immune tolerance in patients with
Smith, H., Shapiro, A. et al. (1998) A randomized, hemophilia and antibodies to factor VIII by combined
double-blind comparison of two dosage levels of treatment with intravenous IgG, cyclophosphamide,
recombinant factor VIIa in the treatment of joint, and factor VIII. N Engl J Med 318: 947–950.
muscle and mucocutaneous haemorrhages in persons
with haemophilia A and B, with and without inhibitors. Oldenburg, J., Picard, J., Schwaab, R., Brackmann, H.,
rFVIIa Study Group. Haemophilia 4: 790–798. Tuddenham, E. and Simpson, E. (1997) HLA
genotype of patients with severe haemophilia A due
Maclean, P., Richards, M., Williams, M., Collins, P., to intron 22 inversion with and without inhibitors of
Liesner, R., Keeling, D. et al. (2011) Treatment factor VIII. Thromb Haemost 77: 238–242.
related factors and inhibitor development in children
with severe haemophilia A. Haemophilia 17: Oldenburg, J., Schroder, J., Brackmann, H., Muller-
282–287. Reible, C., Schwaab, R. and Tuddenham, E. (2004)

70 http://tah.sagepub.com
C Witmer and G Young

Environmental and genetic factors influencing inhibitor development in children with haemophilia A: a case-
development. Semin Hematol 41(1 Suppl. 1): 82–88. control study. Br J Haematol 130: 422–427.
Pavlova, A., Delev, D., Lacroix-Desmazes, S., Santos, A., Annichino-Bizzacchi, J. and Ozelo, M.
Schwaab, R., Mende, M., Fimmers, R. et al. (2009) Inhibitors of factor VIII in hemophilia. N Engl
(2009) Impact of polymorphisms of the major J Med 361: 309–310; author reply 310.
histocompatibility complex class II, interleukin-10,
Scandella, D., Mattingly, M., de Graaf, S. and
tumor necrosis factor-alpha and cytotoxic
Fulcher, C. (1989) Localization of epitopes for human
T-lymphocyte antigen-4 genes on inhibitor
factor VIII inhibitor antibodies by immunoblotting
development in severe hemophilia A. J Thromb
and antibody neutralization. Blood 74: 1618–1626.
Haemost 7: 2006–2015.
Scharrer, I., Bray, G. and Neutzling, O. (1999)
Peyvandi, F., Lotta, L. and Mannucci, P. (2009)
Incidence of inhibitors in haemophilia A patients – a
Inhibitors of factor VIII in hemophilia. N Engl J Med
review of recent studies of recombinant and plasma-
361: 309; author reply 310.
derived factor VIII concentrates. Haemophilia 5:
Qadura, M., Othman, M., Waters, B., Chegeni, R., 145–154.
Walker, K., Labelle, A. et al. (2008) Reduction of the
Schneiderman, J., Nugent, D. and Young, G. (2004)
immune response to factor VIII mediated through
Sequential therapy with activated prothrombin
tolerogenic factor VIII presentation by immature
complex concentrate and recombinant factor VIIa
dendritic cells. J Thromb Haemost 6: 2095–2104.
in patients with severe haemophilia and inhibitors.
Qadura, M., Waters, B., Burnett, E., Chegeni, R., Haemophilia 10: 347–351.
Bradshaw, S., Hough, C. et al. (2009) Recombinant Shapiro, A., Gilchrist, G., Hoots, W., Cooper, H. and
and plasma-derived factor VIII products induce Gastineau, D. (1998) Prospective, randomised trial
distinct splenic cytokine microenvironments in of two doses of rFVIIa (NovoSeven) in haemophilia
hemophilia A mice. Blood 114: 871–880. patients with inhibitors undergoing surgery. Thromb
Ragni, M., Ojeifo, O., Feng, J., Yan, J., Hill, K., Haemost 80: 773–778.
Sommer, S. et al. (2009a) Risk factors for inhibitor Sharathkumar, A., Lillicrap, D., Blanchette, V.,
formation in haemophilia: a prevalent case-control Kern, M., Leggo, J., Stain, A. et al. (2003) Intensive
study. Haemophilia 15: 1074–1082. exposure to factor VIII is a risk factor for inhibitor
Ragni, M., Wu, W., Liang, X., Hsieh, C., Cortese- development in mild hemophilia A. J Thromb Haemost
Hassett, A. and Lu, L. (2009b) Factor VIII-pulsed 1: 1228–1236.
dendritic cells reduce anti-factor VIII antibody Sjamsoedin, L., Heijnen, L., Mauser-Bunschoten, E.,
formation in the hemophilia A mouse model. Exp van Geijlswijk, J., van Houwelingen, H., van Asten,
Hematol 37: 744–754. P. et al. (1981) The effect of activated prothrombin-
complex concentrate (FEIBA) on joint and muscle
Rawle, F., Pratt, K., Labelle, A., Weiner, H.,
bleeding in patients with hemophilia A and antibodies
Hough, C. and Lillicrap, D. (2006) Induction of
to factor VIII. A double-blind clinical trial. N Engl J
partial immune tolerance to factor VIII through prior
Med 305: 717–721.
mucosal exposure to the factor VIII C2 domain.
J Thromb Haemost 4: 2172–2179. Soucie, J., Evatt, B. and Jackson, D. (1998)
Occurrence of hemophilia in the United States. The
Rivard, G., Lillicrap, D., Poon, M., Demers, C., Hemophilia Surveillance System Project Investigators.
Lepine, M., St-Louis, J. et al. (2005) Can activated Am J Hematol 59: 288–294.
recombinant factor VII be used to postpone the
exposure of infants to factor VIII until after 2 years of Su, R., Epp, A., Latchman, Y., Bolgiano, D.,
age? Haemophilia 11: 335–339. Pipe, S. and Josephson, N. (2010) Suppression of
FVIII inhibitor formation in hemophilic mice by
Robbins, D., Kulkarni, R., Gera, R., Scott-Emuakpor, A., delivery of transgene modified apoptotic fibroblasts.
Bosma, K. and Penner, J. (2001) Successful treatment Mol Ther 18: 214–222.
of high titer inhibitors in mild hemophilia A with
avoidance of factor VIII and immunosuppressive Sultan, Y. (1992) Prevalence of inhibitors in a
therapy. Am J Hematol 68: 184–188. population of 3435 hemophilia patients in France.
French Hemophilia Study Group. Thromb Haemost
Saint-Remy, J., Lacroix-Desmazes, S. and Oldenburg, 67: 600–602.
J. (2004) Inhibitors in haemophilia: pathophysiology.
Haemophilia 10(Suppl. 4): 146–151. ter Avest, P., Fischer, K., Mancuso, M., Santagostino, E.,
Yuste, V., van den Berg, H. et al. (2008) Risk stratification
Santagostino, E., Mancuso, M., Rocino, A., for inhibitor development at first treatment for severe
Mancuso, G., Mazzucconi, M., Tagliaferri, A. hemophilia A: a tool for clinical practice. J Thromb
et al. (2005) Environmental risk factors for inhibitor Haemost 6: 2048–2054.

http://tah.sagepub.com 71
Therapeutic Advances in Hematology 4 (1)

van der Bom, J., Mauser-Bunschoten, E., Fischer, K. subcommittee on factor VIII and factor IX of the
and van den Berg, H. (2003) Age at first treatment scientific and standardization committee of the
and immune tolerance to factor VIII in severe International Society on Thrombosis and Haemostasis.
hemophilia. Thromb Haemost 89: 475–479. Thromb Haemost 85: 560.
Verbruggen, B., Novakova, I., Wessels, H., Wight, J. and Paisley, S. (2003) The epidemiology
Boezeman, J., van den Berg, M. and Mauser- of inhibitors in haemophilia A: a systematic review.
Bunschoten, E. (1995) The Nijmegen modification Haemophilia 9: 418–435.
of the Bethesda assay for factor VIII:C inhibitors:
Yang, G., Yao, L. and Lu, Z. (2009) Inhibitors of
improved specificity and reliability. Thromb Haemost
factor VIII in hemophilia. N Engl J Med 361: 309;
73: 247–251.
author reply 310.
Viel, K., Ameri, A., Abshire, T., Iyer, R., Watts, R.,
Lutcher, C. et al. (2009) Inhibitors of factor VIII in black Young, G., Auerswald, G., Jimenez-Yuste, V.,
patients with hemophilia. N Engl J Med 360: 1618–1627. Konkle, B., Lambert, T., Morfini, M. et al. (2011)
When should prophylaxis therapy in inhibitor
Vlot, A., Wittebol, S., Strengers, P., Turenhout, E., patients be considered? Haemophilia 17:
Voorberg, J., van den Berg, H. et al. (2002) Factor e849–e857.
VIII inhibitor in a patient with mild haemophilia A
and an Asn618-->Ser mutation responsive to immune Young, M., Inaba, H., Hoyer, L., Higuchi, M.,
tolerance induction and cyclophosphamide. Br J Kazazian, H., Jr, and Antonarakis, S. (1997)
Haematol 117: 136–140. Partial correction of a severe molecular defect in
hemophilia A, because of errors during expression
Waters, B. and Lillicrap, D. (2009) The molecular of the factor VIII gene. Am J Hum Genet 60:
mechanisms of immunomodulation and tolerance 565–573.
induction to factor VIII. J Thromb Haemost 7: 1446–1456.
Zakarija, A., Harris, S., Rademaker, A., Brewer,
Visit SAGE journals online White, G., Rosendaal, F. 2nd, Aledort, L., Lusher, J., J., Krudysz-Amblo, J., Butenas, S. et al. (2011)
http://tah.sagepub.com
Rothschild, C. and Ingerslev, J. (2001) Definitions Alloantibodies to factor VIII in haemophilia.
SAGE journals in hemophilia. Recommendation of the scientific Haemophilia 17: 636–640.

72 http://tah.sagepub.com

You might also like