You are on page 1of 8

Genetic Risk Factors and Inhibitor Development

in Hemophilia: What Is Known and Searching for


the Unknown
Maurizio Margaglione, MD1 Mariano Intrieri, MD2

1 Medical Genetics, Department of Clinical and Experimental Address for correspondence Maurizio Margaglione, MD, Medical
Medicine, University of Foggia, Foggia, Italy Genetics, Department of Clinical and Experimental Medicine,
2 Department of Medicine and Health Sciences “V. Tiberio,” University University of Foggia, Viale Pinto, Foggia 71122, Italy
of Molise, Campobasso, Italy (e-mail: m.margaglione@unifg.it).

Semin Thromb Hemost

Downloaded by: State University of New York at Stony Brook. Copyrighted material.
Abstract Over the past few decades, important knowledge on why inhibitors develop and better
information about significant risk factors have become available. A series of both
genetic and nongenetic factors are recognized and clinical score systems were
proposed to quantify the risk for each patient. In addition, modulation of the
immunological response was acknowledged to play a pivotal role in the occurrence
of inhibitors. However, with the exception of mutation testing in severe hemophilia B
patients, no single risk factor or clinical score is currently utilized in clinical practice.
Keywords “Omics” technologies are large-scale hypothesis-generating approaches, which pro-
► hemophilia vide the tools to study issues contributing to a complex and multifactorial phenom-
► genetics enon, such as inhibitor development. Newer cutting edge technologies may enable a
► inhibitors more accurate estimation of the personal risk profile and provide a reliable tool to
► variants accurately measure the risk periodically, thereby enabling strategies to foresee and
► genomics prevent inhibitor formation.

Adequate therapy for both hemophilia A (HA) and B (HB) has serious and challenging complication in treating patients
been available for the past four decades. Therapy typically with hemophilia. Inhibitors compromise treatment with
consists of coagulation-factor replacement administered plasma-derived clotting factors or recombinant products
every 2 to 3 days, either prophylactically (i.e., to prevent by neutralizing FVIII and FIX activity, and will preclude
bleeding episodes) or episodically (on demand when bleed- such treatment in the case of high inhibitor levels. During
ing occurs). Such replacement treatment has led to consider- their lifetime, the incidence of inhibitors is 20 to 30% in
able improvements in the life expectancy and physical status severe HA and 3 to 5% in HB patients.2 Because of the higher
of patients.1 At present, the quality of their life is similar to levels of morbidity and mortality and a worse quality of life,
that of the general population and this goal can be achieved attempts to restore the efficacy of replacement therapy are
with adequate replacement therapy, using either recombi- mandatory. Why most patients are tolerant to the “foreign”
nant products or plasma-derived clotting factors that protein being administered, whereas some patients develop
undergo extensive purification and viral inactivation. neutralizing antibodies, is far from clear and it is a matter of
The development of alloantibodies (inhibitors) against considerable discussion.
therapeutically administered factor VIII (FVIII) or factor IX The clinical and economic burden of the management of
(FIX) impairs the effectiveness of treatment, and is the most patients with persistent inhibitors fostered substantial

Issue Theme Alloantibodies and Copyright © by Thieme Medical DOI https://doi.org/


Congenital Bleeding Disorders: New Publishers, Inc., 333 Seventh Avenue, 10.1055/s-0038-1660816.
Insights in the Pathogenesis and New York, NY 10001, USA. ISSN 0094-6176.
Management; Guest Editors: Antonio Tel: +1(212) 584-4662.
Coppola, MD, Massimo Franchini, MD,
Cristina Santoro, MD, PhD, and Annarita
Tagliaferri, MD.
Inhibitor Development as a Multifactorial Event Margaglione, Intrieri

research effort to elucidate the etiology of inhibitors and the A2 and C2 domains.6 The C2 domain contains binding
several risk factors for their occurrence have been identified. sites to von Willebrand factor and phosphatidylserine on
This effort allowed for increased knowledge on the genetic activated platelets. Specific F8 variants, such as Arg593Cys in
and environmental determinants of the risk of inhibitor the A2 and Trp2229Cys in the C2 domain, are highly related to
development and the identification of predictors of devel- inhibitor development. On the other hand, replacement
oping inhibitors. therapy in patients completely lacking FVIII may lead to
Notwithstanding, the identification of risk factors immunization with the “foreign” protein and cause the
(►Table 1) remains crucial to optimize candidate selection development of autoantibodies. Indeed, mutations predict-
to individualized treatment regimens or modification of ing a null allele develop an inhibitor more frequently (24–
immunological factors according to the predicted risk and 71%), compared with missense mutations (5%). These find-
might be of help for the detection of strategies for the ings support the hypothesis that trace amounts of FVIII
prevention of inhibitors and a decrease in the total incidence protein, albeit nonfunctional, which are synthesized are
of inhibitors among hemophilia patients. sufficient to induce immune tolerance in most HA patients.
In addition, mutations at specific regions of F8 (hot spots,
exons coding for the heavy chain) are associated with a lower
Nonmodifiable Risk Factors
risk of developing inhibitors. Indeed, the risk of inhibitor
Genotype formation was similar in patients carrying the inversion of

Downloaded by: State University of New York at Stony Brook. Copyrighted material.
HA is an X-linked bleeding disorder caused by heterogeneous the intron 22 when compared with those with intron 1
mutations in FVIII gene (F8). The incidence of HA is estimated inversion (odds ratio [OR] 0.9; 95% confidence interval
to be between 1:5,000 and 1:10,000 in men.3 F8 maps to the [CI]: 0.6–1.5) and splice site mutations (OR: 1.0; 95% CI:
distal end of the long arm of the X-chromosome (Xq28) and 0.6–1.5), but significantly higher in those with large dele-
spans 186 kb of genomic DNA. It consists of 26 exons and tions and nonsense mutations (OR: 3.6; 95% CI: 2.3–5.7), and
encodes a mature protein of 2,332 amino acids.4 Over the last lower in HA patients carrying small deletions/insertions and
few decades, rapidly increasing numbers of causative gene missense mutations (OR: 0.5; 95% CI: 0.4–0.6).5
alterations have been described in different ethnic groups. At It is well known that the degree of severity of the disease
present, more than 900 mutations within the F8 coding and represents an important factor for the occurrence of inhi-
untranslated regions have been identified and listed in the F8 bitor. At variance with severe patients, the inhibitor risk is 5
variant database (URL: http://www.factorviii-db.org). These to 10% in mild to moderate HA.7,8 Of note, severe and
findings support the concept that HA has a very high muta- nonsevere HAs display significant differences in prevalence
tional heterogeneity and represents a challenge for the of type of causative mutations.9 Severe and nonsevere HAs
provision of genetic services because carrier and prenatal are also likely to be associated with different timing in risk of
diagnosis cannot be based on the screening of a limited inhibitor development.8
number of common mutations. Similar to HA, HB is an X-linked bleeding disorder caused
The type of F8 mutation is one of the most important risk by heterogeneous mutations in the FIX gene (F9). The inci-
factors predisposing to inhibitor development in HA.5 In HA dence of HB is estimated to be 1:25,000 in men.3 F9 maps to
patients, the majority of the inhibitors are directed against the distal end of the long arm of the X-chromosome (Xq27),

Table 1 Established and proposed risk factors for inhibitor development

Disease Risk factor Level of evidence


HA Nonmodifiable F8 gene mutation Well established
Family history Well established
Ancestry Established
HLA alleles Weak
IL-10 common alleles Some evidence
TNF-α common alleles Some evidence
CTLA4 common alleles Some evidence
ABO blood group Weak
Modifiable Traumatic injury or surgical challenges Some evidence
High dosage and/or prolonged treatment at the beginning of the therapy Some evidence
Primary prophylaxis vs. on-demand treatment Some evidence
Recombinant products vs. plasma-derived high purity concentrates Evidence
HB Nonmodifiable F9 gene mutation Well established

Abbreviations: HA, hemophilia A; HB, hemophilia B.

Seminars in Thrombosis & Hemostasis


Inhibitor Development as a Multifactorial Event Margaglione, Intrieri

very close to F8, and spans 34 kb of genomic DNA. F9 consists gested to explain this association.28 However, at variance
of 8 exons and encodes a mature protein of 461 amino acids.4 with this hypothesis, patients of African/Hispanic ancestry
At variance with patients with HA, those with HB can with the H1 haplotype still had significantly higher inhibitor
experience anaphylactic reactions to FIX concentrates at frequencies than Caucasians, suggesting that the mechan-
the time of inhibitor development.10 The smaller molecular isms accounting for these ancestry differences remain
size of FIX compared with FVIII, allowing for a rapid diffusion unclear.29
into extravascular spaces, and the higher amounts of exo-
genous protein given to HB patients, due to physiologically Genes Encoding Immune-Regulatory Cytokines
higher plasma levels of FIX (5 µg/mL) than FVIII (0.1 µg/mL) As demonstrated in autoimmune disease, defects in genes
may, in part, explain reasons for anaphylactic response.11 leading to an altered function of cytokines or their receptors
Although there have been occasional reports of large dele- may cause an imbalance of immune response. Recently,
tions, HB is usually associated with hundreds of different Astermark et al found a strong association between poly-
missense, nonsense, frameshifts, and splicing mutations.12 morphisms in the promoter regions of the interleukin-10
These gene variants frequently result in the production of a (IL10), tissue necrosis factor (TNF)-α, and the cytotoxic T-
defective, nonfunctioning, but immunologically detectable lymphocyte antigen 4 (CTLA4) genes and the development of
FIX in the plasma of HB patients (cross-reacting material FVIII inhibitors.30–32 These genes are known to be involved in
positive [CRMþ]). Individuals with large gene deletions, the regulation of antibody responses and could, therefore, be

Downloaded by: State University of New York at Stony Brook. Copyrighted material.
frameshift, and nonsense mutations are usually CRM and of major importance in the development of FVIII inhibitors.
are most susceptible to the development of inhibitors against The CTLA4 protein is a surface molecule expressed on
FIX, accounting approximately for 70% of cases, while mis- activated T cells and plays a critical role as a negative
sense mutations, which constitute the majority of genotypes regulator of T cell activation. Some polymorphisms of the
in HB, are at very low risk of inhibitor formation.13–15 The CTLA4 gene (promoter 318 C/T and exon 1 þ 49 A/G) are
increased risk of inhibitors in patients carrying large gene associated with different levels of expression of this mole-
deletions, frameshift, and nonsense mutations has prompted cule on the cell surface. A significant protective association
recommending mutation testing in severe HB patients to between the inhibitor development and the 318 T allele,
identify who are at risk for inhibitor development and but not the þ49 A/G polymorphism, has been found.33
anaphylactic reactions.16 In addition, entity of risk in pre- IL-10 is an important cytokine with a broad spectrum of
viously untreated severe HB patients is poorly known, as biological activities including immunosuppressive, anti-
most studies do not provide specific information and that inflammatory, and B cell stimulating properties. An
factors other than genotype have not been studied in such increased inhibitor risk has been shown in patients with a
patients.17,18 Thus, information reported in the following microsatellite polymorphism in the promoter of the IL-10
paragraphs refer to HA. gene compared with noncarriers.30
TNF-α is a proinflammatory cytokine that plays a key role
Family History in the pathogenesis of many infectious and inflammatory
Family history is also a risk factor associated with the diseases. Many studies have investigated the TNF-α poly-
development of inhibitors.19–21 In a study, in which 249 morphisms in autoimmune diseases in which the dysregula-
families with severe HA containing two or more siblings tion of TNF-α production might have played a role. In
were evaluated, a high concordance between siblings (78.3%) particular, a SNP in the promoter region, 308 A/G, is
and a significant relative risk (3.2; 95% CI: 2.1–4.9) for the relevant: the TNF-α 308 A allele has been found to correlate
development of an inhibitor was found.20 Noteworthy, with enhanced spontaneous and stimulated TNF-α produc-
although higher than that observed in non-twin siblings, tion. The polymorphism is associated with antibody forma-
concordance was not absolute (90%) even in monozygotic tion in patients with myasthenia gravis.24 A similar
twins.22 On the other hand, concordance in families with association with the occurrence of inhibitors has been shown
inhibitors was 42%, and 72% of these inhibitors had the same in a subgroup of patients with severe HA enrolled in the
anamnestic (high-responding) features.21 Thus, the lack of a Mälmo International Brother Study (MIBS).33 The potential
complete concordance between siblings (all or none with a role of these polymorphic markers is further supported by
history of inhibitors) shows that this genetic susceptibility their distinct distribution in different populations.33 Finally,
could not be predicted by the F8 mutation itself. significant associations with the risk for inhibitor formation
were found with haplotypes at various loci of the IL-10, IL-1,
Ancestry IL-2, and IL-12 genes.34 However, a more comprehensive
It is well known that HA patients of African or Hispanic genetic approach enlisting a large number of brother pairs
heritage have an increased risk of inhibitor develop- discordant for inhibitor status did not confirm a pivotal effect
ment.20,23–26 This association was recently confirmed in a of CTLA4, IL-10, and TNF-α gene polymorphisms.35
study that controlled for the F8 genotype.27 Differences in
amino acids between currently available replacement pro- Other Loci
ducts (which are all from strains harboring F8 H1 and H2 The ABO blood group has been suggested to significantly
haplotypes), and patients of African/Hispanic ancestry (more modulate the appearance of inhibitors. The relative risk in O
commonly carrying H3 or H4 haplotypes), have been sug- versus non-O blood group was 0.55 (95% CI: 0.33–0.92) and

Seminars in Thrombosis & Hemostasis


Inhibitor Development as a Multifactorial Event Margaglione, Intrieri

0.40 (95% CI: 0.21–0.77) for any and for high-titer inhibitors, decreased overall inhibitor incidence after 20 days of expo-
respectively.36 Because the glycosyltransferase activity, sure to exogenous FVIII. The association was more evident in
which is encoded by the ABO gene, also significantly affects severe HA patients with low-risk than in those with high-risk
FVIII and its carrier, von Willebrand factor, the differences in F8 genotypes.47
posttranslational glycosylation may provide an intriguing
explanation for the differential risk. The HLA locus has been Type of Factor Concentrate
suggested as another important genetic determinant capable Purified FVIII products were available in the 1970s, whereas
of influencing the occurrence of inhibitors in HA.33,37 HLA recombinant FVIII products became available in the early
class II molecules mediate the processing of antigenic extra- 1990s. The retrospective evaluation of the incidence of
cellular proteins, such as the exogenous FVIII administered to inhibitors in patients treated with plasma-derived high
HA patients. Findings from a series of studies showed a purity concentrate, first generation, and second recombinant
significant association between the HLA system, in particular products suggested an increased incidence of inhibitors
HLA class II molecules, and the development of inhibitors. using later products. This increased immunogenicity is
However, the strength of this association varies throughout hypothesized to be secondary to alterations in posttransla-
the world and depends on the genetic background of the tional modifications of FVIII and a reduction of von Will-
different groups of population investigated.38 ebrand factor binding.48–51 These findings were challenged
by two systematic reviews52,53 and a patient-level meta-

Downloaded by: State University of New York at Stony Brook. Copyrighted material.
Final Considerations analysis.46 Then, a series of ad hoc analyses of national
Although it is well known that a genetic predisposition to untreated HA cohorts54–56 showed a significant difference
inhibitor development to FVIII/FIX proteins exists, it is also of inhibitor incidence among recombinant products. This
clear that the classification into high-risk and low-risk evidence was recently supported by findings from a national
mutations is likely an oversimplification. From a clinical cohort.57 The only randomized trial so far available strongly
point of view, to better define the individual risk for the supported the concept of a higher inhibitor incidence among
occurrence of inhibitors in newly diagnosed patients starting untreated patients using recombinant products.58 On the
replacement treatment, it would be helpful to also consider other hand, the current evidence does not show this
the inhibitor family history and ancestry, which may both increased risk in patients after the first 50 to 75 exposure
include other genetic determinants. days, suggesting that factors leading to inhibitors develop-
ment are likely to play their role only early during the first
exposure days.
Possible Modifiable Risk Factors
Treatment Related Final Considerations
Although the large proportion of patients who develop Overall, available data are fairly consistent and suggest that a
inhibitors have severe disease, inhibitors can also occur in series of potentially modifiable risk factors exist that can be
patients with mild and moderate hemophilia. Likewise, modulated with the aim of lowering the occurrence of
inhibitors are most common among previously untreated inhibitors.
patients and the risk decreases as the number of exposure
days rises. Inhibitors can also occur in patients who have
The Multifactorial Model
been already exposed for more than 50 days. However, in this
clinical setting, the overall rate of inhibitors is low.39 More- Since its introduction, the multifactorial threshold model has
over, inhibitor development often occurs in association with been widely applied in genetic counselling for qualitative
traumatic injury or surgical challenges that result in signifi- (discrete) traits.59–61 The multifactorial model includes the
cant and prolonged exposures to replacement factor con- influence of many factors, genetic and environmental. The
centrates. The presentation to the immune system of the risk does not change drastically with the presence or the
exogenous FVIII alone may not be sufficient for initiating an absence of one of them. This implies that the susceptibility to
immune response. The intensity of the FVIII exposure has inhibitor formation is largely accounted for by the clustering
been suggested to be a risk factor for inhibitor formation of several, possibly inherited, risk factors. Usually, geneticists
because significant cell injury or inflammation leads to admit that in a multifactorial phenotype, several loci, but not
immunologic “danger signals” that stimulate antigen-pre- an unlimited number, are involved in the expression of the
senting cells and upregulate an immunologic response trait; there is no classical (Mendelian) dominance or reces-
against exogenous FVIII, which could promote inhibitor siveness at each of these loci; the loci act in concert in an
development.40–42 Several studies have shown a possible additive fashion, each adding or detracting a small amount
link between intensity (high dosage and/or prolonged treat- from the phenotype, and the environment interacts with the
ment) of early treatment and increased risk of inhibitor genotype to produce the final phenotype. None of these
development.25,43–45 This prompted the concept that pri- factors is either necessary or sufficient for the occurrence
mary prophylaxis has a significantly lower inhibitor rate as of inhibitors, but the totality makes inhibitor development
compared with patients treated on demand. The hypothesis more likely. This means that the liability to develop an
was confirmed in a recent patient-level meta-analysis.46 inhibitor-prone phenotype is continuously distributed in
However, prophylaxis seems to be associated with a the HA population because of the additive effects of genetic

Seminars in Thrombosis & Hemostasis


Inhibitor Development as a Multifactorial Event Margaglione, Intrieri

and environmental factors. Our current thinking is that only allows for a timely estimation of the individual risk when an
in individuals whose liability exceeds a certain threshold will incidental risk factor happens.
inhibitors then occur. However, because the phenotype alone
may not be able to distinguish different subgroups, genetic
Looking Forward
heterogeneity may affect the ability to identify genetic risk
factors associated with inhibitor formation. In recent years, new high-throughput technologies for assay-
On the basis of the above-mentioned model, the ing biological activity using holistic approaches for biomo-
advancement in the understanding of risk factors that lecules such as DNA, RNA, and proteins have been developed,
underlie the occurrence of inhibitors fostered the search where no hypothesis is known or proposed, but all data are
for the creation of a clinical scoring system to quantify the acquired and analyzed to generate hypotheses. Such large-
risk of each patient. In HA, a clinical scoring system using scale approaches have enabled unprecedented views of
three factors (F8 gene mutation, family history of inhibitor, biological systems at the molecular level (►Fig. 1). The fields
and intensive treatment at initial exposure) to predict of research associated with obtaining and understanding
inhibitor formation identified that patients without any such measurements—for instance, genomics, transcrip-
risk factors (risk score ¼ 0 points) had an inhibitor inci- tomics, and proteomics—are sometimes referred to in aggre-
dence of 6%, whereas those with two or more than three gate as “omics.” As a whole, “omics” technologies allow for a
points showed an incidence of 23 and 57%, respectively.62 A much more powerful approach to unravel key regulators

Downloaded by: State University of New York at Stony Brook. Copyrighted material.
different clinical scoring system showed rates ranging from leading to multifactorial conditions such as inhibitors
4% in the most favorable combination (O blood type, F8 low development.
risk mutation, and use of plasma-derived FVIII products) to Genome-wide expression profiling studies investigate the
nearly 70% (non-O blood type, high-risk mutation, and use activity of the genome rather than the inherent genome
of recombinant FVIII products).36 Finally, a 15-item predic- variation and may identify which genes are “switched on”
tion score model using a 5-point linear scale (with “1” being and how much in any given situation such as when inhibitors
very unlikely and “5” very likely) found important factors occur. In patients with HA, the CD4þ T cells–mediated
for inhibitor development, including genetic factors (mean immunologic response is strongly related to the synthesis
score: 4.50) and early intensive treatment (mean score: of anti-FVIII antibodies.64 In the immunological response,
4.07), as well as important management strategies to activation of CD4þ T cells regulates the proliferation and
reduce inhibitor development, including early onset of differentiation of B cells and results in the acquisition of a
prophylaxis (mean score: 3.54) and avoidance of elective well-organized expression of multiple effector genes. Tran-
surgery (mean score: 4.32).63 The retrospective nature of scriptional profiling could be investigated to assess
the analysis hardly hampers the use of these clinical scoring responses of CD4þ T cells, especially type-2 helper T cells,
systems. In addition, none of the clinical scoring systems to different exogenous formulations of FVIII and identify

Fig. 1 Schematic representation of most represented “omics” technologies.

Seminars in Thrombosis & Hemostasis


Inhibitor Development as a Multifactorial Event Margaglione, Intrieri

cell-specific responses which could predict inhibitor time, depending on the appearance of transient risk factors.
development.65 If it will become possible to have a reliable tool to accurately
It is well known that gene expression is modulated by measure from time to time the risk, it will become realistic to
mechanisms other than the DNA sequence. Epigenetics have strategies to foresee and prevent inhibitor formation.
refers to dynamic changes in chromatin structure that play
a key role in regulating genome functions, including tran- Conflict of Interest
scription. On the whole, mechanisms such as DNA methyla- None.
tion and posttranslational modifications of histone proteins
play a significant role in the regulation of inducible immune-
responsive gene transcription. In CD4þ T cells, genes with References
different basal expression levels are associated with specific 1 Remor E, Young NL, Von Mackensen S, Lopatina EG. Disease-
combinations of histone modifications and reorganization at specific quality-of-life measurement tools for haemophilia
gene promoters and enhancers in response to T cell activa- patients. Haemophilia 2004;10(Suppl 4):30–34
tion.66 Thus, cells depending on the phase of their life possess 2 Dimichele D. The North American Immune Tolerance Registry:
contributions to the thirty-year experience with immune toler-
distinct and characteristic patterns, “signatures,” of gene
ance therapy. Haemophilia 2009;15(01):320–328
expression and chromatin structural adaptation. 3 Skinner MW. WFH: closing the global gap–achieving optimal
In general, genes with a low expression rate are related to

Downloaded by: State University of New York at Stony Brook. Copyrighted material.
care. Haemophilia 2012;18(Suppl 4):1–12
promoters with repressed chromatin signatures, whereas an 4 Bowen DJ. Haemophilia A and haemophilia B: molecular insights.
active chromatin signature shows intermediate to high gene Mol Pathol 2002;55(02):127–144
expression levels.67 In HA, understanding how the exposure 5 Gouw SC, van den Berg HM, Oldenburg J, et al. F8 gene mutation
type and inhibitor development in patients with severe hemo-
to FVIII modulates the dynamic interplay between hetero-
philia A: systematic review and meta-analysis. Blood 2012;119
chromatin and euchromatin formation leading to chromatin (12):2922–2934
reorganization and, in turn, to the regulation of gene expres- 6 Key NS. Inhibitors in congenital coagulation disorders. Br J
sion is expected to be crucial for the identification of key Haematol 2004;127(04):379–391
mechanisms for the occurrence of inhibitors. Thus, a combi- 7 Oldenburg J, Pavlova A. Genetic risk factors for inhibitors to
factors VIII and IX. Haemophilia 2006;12(Suppl 6):15–22
nation of transcriptional and epigenetic profiling to study
8 Eckhardt CL, van Velzen AS, Peters M, et al; INSIGHT Study Group.
both gene expression and chromatin signatures, together
Factor VIII gene (F8) mutation and risk of inhibitor development
with the additional help of in silico information from bioin- in nonsevere hemophilia A. Blood 2013;122(11):1954–1962
formatics, computational modelling, and pathway analysis, 9 Margaglione M, Castaman G, Morfini M, et al; AICE-Genetics
will boost our knowledge of how inhibitors develop and in Study Group. The Italian AICE-Genetics hemophilia A database:
response to which stimuli. results and correlation with clinical phenotype. Haematologica
2008;93(05):722–728
10 Chitlur M, Warrier I, Rajpurkar M, Lusher JM. Inhibitors in factor
Final Considerations IX deficiency a report of the ISTH-SSC international FIX inhibitor
Cutting edge technologies may enable a thorough and clever registry (1997-2006). Haemophilia 2009;15(05):1027–1031
estimation of the personal risk profile, which is currently 11 Warrier I, Lusher JM. Development of anaphylactic shock in
relatively inaccurate, involving the assessment of “omics” haemophilia B patients with inhibitors. Blood Coagul Fibrinolysis
signatures or biomarkers. This approach will represent a 1998;9(Suppl 1):S125–S128
12 Rallapalli PM, Kemball-Cook G, Tuddenham EG, Gomez K, Per-
welcome improvement in the ability to measure the risk for
kins SJ. An interactive mutation database for human coagulation
inhibitors development in a meaningful way. factor IX provides novel insights into the phenotypes and
genetics of hemophilia B. J Thromb Haemost 2013;11(07):
1329–1340
Conclusion 13 Oldenburg J, Schröder J, Brackmann HH, Müller-Reible C, Schwaab R,
Tuddenham E. Environmental and genetic factors influencing inhibi-
There is good evidence that host genetic factors are impor-
tor development. Semin Hematol 2004;41(01, Suppl 1):82–88
tant, but not the only determinants of the immunologic
14 Belvini D, Salviato R, Radossi P, et al; AICE HB Study Group.
response to FVIII administration. Genetic studies demon- Molecular genotyping of the Italian cohort of patients with
strated that many responses are partially predictable, and hemophilia B. Haematologica 2005;90(05):635–642
modifiable factors modulate the appearance of inhibitors. 15 Saini S, Hamasaki-Katagiri N, Pandey GS, et al. Genetic determi-
The comprehension of mechanisms leading to inhibitors nants of immunogenicity to factor IX during the treatment of
haemophilia B. Haemophilia 2015;21(02):210–218
formation will pave the way for the complete understanding
16 Rocino A, Coppola A, Franchini M, et al; Italian Association of
of how risk factors determine their development. It can be Haemophilia Centres (AICE) Working Party. Principles of treat-
anticipated that different risk factors may interact each other ment and update of recommendations for the management of
and give raise to an epistatic effect. Indeed, very recently it haemophilia and congenital bleeding disorders in Italy. Blood
was suggested that the use of recombinant products gives a Transfus 2014;12(04):575–598
paradoxical high risk in patients carrying low-risk F8 gene 17 DiMichele D. Inhibitor development in haemophilia B: an orphan
disease in need of attention. Br J Haematol 2007;138(03):
mutations. Thus, it is conceivable that the specific personal
305–315
array of nonmodifiable and possible modifiable risk factors 18 Franchini M, Santoro C, Coppola A. Inhibitor incidence in pre-
may identify a unique risk profile for each patient with viously untreated patients with severe haemophilia B: a systema-
hemophilia. The personal risk profile may change over tic literature review. Thromb Haemost 2016;116(01):201–203

Seminars in Thrombosis & Hemostasis


Inhibitor Development as a Multifactorial Event Margaglione, Intrieri

19 Gill JC. The role of genetics in inhibitor formation. Thromb Hae- 37 Hay CR, Ollier W, Pepper L, et al; UKHCDO Inhibitor Working
most 1999;82(02):500–504 Party. HLA class II profile: a weak determinant of factor VIII
20 Astermark J, Berntorp E, White GC II, Kroner BL; MIBS Study inhibitor development in severe haemophilia A. Thromb Haemost
Group. The Malmö International Brother Study (MIBS): further 1997;77(02):234–237
support for genetic predisposition to inhibitor development in 38 Wieland I, Wermes C, Eifrig B, et al. Inhibitor-Immunology-Study.
hemophilia patients. Haemophilia 2001;7(03):267–272 Different HLA-types seem to be involved in the inhibitor devel-
21 Astermark J, Oldenburg J, Escobar M, White GC II, Berntorp E; opment in haemophilia A. Hamostaseologie 2008;28(Suppl 1):
Malmö International Brother Study study group. The Malmö S26–S28
International Brother Study (MIBS). Genetic defects and inhibitor 39 Xi M, Makris M, Marcucci M, Santagostino E, Mannucci PM, Iorio
development in siblings with severe hemophilia A. Haematolo- A. Inhibitor development in previously treated hemophilia A
gica 2005;90(07):924–931 patients: a systematic review, meta-analysis, and meta-regres-
22 Klintman J, Hillarp A, Donfield S, Berntorp E, Astermark J. Anti- sion. J Thromb Haemost 2013;11(09):1655–1662
body formation and specificity in Bethesda-negative brother pairs 40 Gallucci S, Matzinger P. Danger signals: SOS to the immune
with haemophilia A. Haemophilia 2013;19(01):106–112 system. Curr Opin Immunol 2001;13(01):114–119
23 Aledort LM, Dimichele DM. Inhibitors occur more frequently in 41 Matzinger P. The danger model: a renewed sense of self. Science
African-American and Latino haemophiliacs. Haemophilia 1998; 2002;296(5566):301–305
4(01):68 42 Kono H, Rock KL. How dying cells alert the immune system to
24 Scharrer I, Bray GL, Neutzling O. Incidence of inhibitors in danger. Nat Rev Immunol 2008;8(04):279–289
haemophilia A patients–a review of recent studies of recombi- 43 Gouw SC, van den Berg HM, le Cessie S, van der Bom JG.

Downloaded by: State University of New York at Stony Brook. Copyrighted material.
nant and plasma-derived factor VIII concentrates. Haemophilia Treatment characteristics and the risk of inhibitor development:
1999;5(03):145–154 a multicenter cohort study among previously untreated patients
25 Ragni MV, Ojeifo O, Feng J, et al; Hemophilia Inhibitor Study. Risk with severe hemophilia A. J Thromb Haemost 2007;5(07):
factors for inhibitor formation in haemophilia: a prevalent case- 1383–1390
control study. Haemophilia 2009;15(05):1074–1082 44 Gouw SC, van der Bom JG, Marijke van den Berg H. Treatment-
26 Carpenter SL, Michael Soucie J, Sterner S, Presley R; Hemophilia related risk factors of inhibitor development in previously
Treatment Center Network (HTCN) Investigators. Increased pre- untreated patients with hemophilia A: the CANAL cohort study.
valence of inhibitors in Hispanic patients with severe haemophi- Blood 2007;109(11):4648–4654
lia A enrolled in the Universal Data Collection database. 45 Maclean PS, Richards M, Williams M, et al; Paediatric Working
Haemophilia 2012;18(03):e260–e265 Party of UKHCDO. Treatment related factors and inhibitor devel-
27 Miller CH, Benson J, Ellingsen D, et al; Hemophilia Inhibitor opment in children with severe haemophilia A. Haemophilia
Research Study Investigators. F8 and F9 mutations in US haemo- 2011;17(02):282–287
philia patients: correlation with history of inhibitor and race/ 46 Marcucci M, Mancuso ME, Santagostino E, et al. Type and intensity
ethnicity. Haemophilia 2012;18(03):375–382 of FVIII exposure on inhibitor development in PUPs with haemo-
28 Viel KR, Ameri A, Abshire TC, et al. Inhibitors of factor VIII in black philia A. A patient-level meta-analysis. Thromb Haemost 2015;
patients with hemophilia. N Engl J Med 2009;360(16):1618–1627 113(05):958–967
29 Schwarz J, Astermark J, Menius ED, et al; Hemophilia Inhibitor 47 Gouw SC, van den Berg HM, Fischer K, et al; PedNet and Research
Genetics Study Combined Cohort. F8 haplotype and inhibitor risk: of Determinants of INhibitor Development (RODIN) Study Group.
results from the Hemophilia Inhibitor Genetics Study (HIGS) Intensity of factor VIII treatment and inhibitor development in
Combined Cohort. Haemophilia 2013;19(01):113–118 children with severe hemophilia A: the RODIN study. Blood 2013;
30 Astermark J, Oldenburg J, Pavlova A, Berntorp E, Lefvert AK; MIBS 121(20):4046–4055
Study Group. Polymorphisms in the IL10 but not in the IL1beta 48 Hironaka T, Furukawa K, Esmon PC, et al. Comparative study of the
and IL4 genes are associated with inhibitor development in sugar chains of factor VIII purified from human plasma and from
patients with hemophilia A. Blood 2006;107(08):3167–3172 the culture media of recombinant baby hamster kidney cells. J Biol
31 Astermark J, Oldenburg J, Carlson J, et al. Polymorphisms in the Chem 1992;267(12):8012–8020
TNFA gene and the risk of inhibitor development in patients with 49 Dasgupta S, Repessé Y, Bayry J, et al. VWF protects FVIII from
hemophilia A. Blood 2006;108(12):3739–3745 endocytosis by dendritic cells and subsequent presentation to
32 Astermark J, Wang X, Oldenburg J, Berntorp E, Lefvert AK; MIBS immune effectors. Blood 2007;109(02):610–612
Study Group. Polymorphisms in the CTLA-4 gene and inhibitor 50 Qadura M, Waters B, Burnett E, et al. Recombinant and plasma-
development in patients with severe hemophilia A. J Thromb derived factor VIII products induce distinct splenic cytokine
Haemost 2007;5(02):263–265 microenvironments in hemophilia A mice. Blood 2009;114(04):
33 Pavlova A, Delev D, Lacroix-Desmazes S, et al. Impact of poly- 871–880
morphisms of the major histocompatibility complex class II, 51 Grancha S, Navajas R, Marañón C, Paradela A, Albar JP, Jorquera JI.
interleukin-10, tumor necrosis factor-alpha and cytotoxic T-lym- Incomplete tyrosine 1680 sulphation in recombinant FVIII con-
phocyte antigen-4 genes on inhibitor development in severe centrates. Haemophilia 2011;17(04):709–710
hemophilia A. J Thromb Haemost 2009;7(12):2006–2015 52 Iorio A, Halimeh S, Holzhauer S, et al. Rate of inhibitor develop-
34 Lozier JN, Rosenberg PS, Goedert JJ, Menashe I. A case-control ment in previously untreated hemophilia A patients treated with
study reveals immunoregulatory gene haplotypes that influence plasma-derived or recombinant factor VIII concentrates: a sys-
inhibitor risk in severe haemophilia A. Haemophilia 2011;17(04): tematic review. J Thromb Haemost 2010;8(06):1256–1265
641–649 53 Franchini M, Coppola A, Rocino A, et al; Italian Association of
35 Astermark J, Donfield SM, Gomperts ED, et al; Hemophilia Inhi- Hemophilia Centers (AICE) Working Group. Systematic review of
bitor Genetics Study (HIGS) Combined Cohort. The polygenic the role of FVIII concentrates in inhibitor development in pre-
nature of inhibitors in hemophilia A: results from the Hemophilia viously untreated patients with severe hemophilia a: a 2013
Inhibitor Genetics Study (HIGS) Combined Cohort. Blood 2013; update. Semin Thromb Hemost 2013;39(07):752–766
121(08):1446–1454 54 Gouw SC, van der Bom JG, Ljung R, et al; PedNet and RODIN Study
36 Franchini M, Coppola A, Mengoli C, et al; Ad Hoc Study Group. Group. Factor VIII products and inhibitor development in severe
Blood group O protects against inhibitor development in severe hemophilia A. N Engl J Med 2013;368(03):231–239
hemophilia A patients. Semin Thromb Hemost 2017;43(01): 55 Calvez T, Chambost H, Claeyssens-Donadel S, et al; FranceCoag
69–74 Network. Recombinant factor VIII products and inhibitor

Seminars in Thrombosis & Hemostasis


Inhibitor Development as a Multifactorial Event Margaglione, Intrieri

development in previously untreated boys with severe hemophi- 61 Smith C. Recurrence risks for multifactorial inheritance. Am J
lia A. Blood 2014;124(23):3398–3408 Hum Genet 1971;23(06):578–588
56 Collins PW, Palmer BP, Chalmers EA, et al; UK Haemophilia Centre 62 ter Avest PC, Fischer K, Mancuso ME, et al; CANAL Study Group.
Doctors’ Organization. Factor VIII brand and the incidence of factor Risk stratification for inhibitor development at first treatment for
VIII inhibitors in previously untreated UK children with severe severe hemophilia A: a tool for clinical practice. J Thromb Hae-
hemophilia A, 2000-2011. Blood 2014;124(23):3389–3397 most 2008;6(12):2048–2054
57 Calvez T, Chambost H, d’Oiron R, et al; for FranceCoag Collabora- 63 van den Berg HM, Chalmers EA. Clinical prediction models for
tors. Analyses of the FranceCoag cohort support differences in inhibitor development in severe hemophilia A. J Thromb Haemost
immunogenicity among one plasma-derived and two recombi- 2009;7(Suppl 1):98–102
nant factor VIII brands in boys with severe hemophilia A. Hae- 64 Qian J, Collins M, Sharpe AH, Hoyer LW. Prevention and treatment
matologica 2018;103(01):179–189 of factor VIII inhibitors in murine hemophilia A. Blood 2000;95
58 Peyvandi F, Mannucci PM, Garagiola I, et al. A randomized trial of (04):1324–1329
factor VIII and neutralizing antibodies in hemophilia A. N Engl J 65 Pratt KP, Thompson AR. B-cell and T-cell epitopes in anti-factor
Med 2016;374(21):2054–2064 VIII immune responses. Clin Rev Allergy Immunol 2009;37(02):
59 Crittenden LB. An interpretation of familial aggregation based on 80–95
multiple genetic and environmental factors. Ann N Y Acad Sci 66 Schones DE, Zhao K. Genome-wide approaches to studying chro-
1961;91:769–780 matin modifications. Nat Rev Genet 2008;9(03):179–191
60 Falconer DS. The inheritance of liability to certain diseases 67 Hon G, Wang W, Ren B. Discovery and annotation of functional
estimated from the incidence among relatives. Ann Hum Genet chromatin signatures in the human genome. PLOS Comput Biol

Downloaded by: State University of New York at Stony Brook. Copyrighted material.
1965;29:51–76 2009;5(11):e1000566

Seminars in Thrombosis & Hemostasis

You might also like