499
Views
5
CrossRef citations to date
0
Altmetric
CURRENT CLINICAL PRACTICE - HEMOPHILIA

Alloantibodies in previously untreated hemophilia A patients: the role of environmental factors

, , &
Pages 183-190 | Published online: 15 Nov 2013

Abstract

The therapy of hemophilia A has nowadays reached a high degree of quality, being probably the most efficacious and safe treatment available among other monogenic disorders. In this context, the most challenging complication of therapy has become the development of inhibitory alloantibodies against FVIII. These inhibitors, which develop in up to 30% of severe hemophilia A patients, render replacement therapies ineffective, limit patient access to a safe and effective standard of care, and predispose them to an increased risk of morbidity and mortality. Several possible risk factors, both genetic and environmental, for inhibitor development have been identified in previously untreated patients, confirming that the etiology of this phenomenon is multifactorial. In this narrative review, we will focus on acquired risk factors, discussing the pathogenic and clinical data available from the literature analysis.

Introduction

The development during the last 25 years of viral inactivation procedures and of recombinant products has greatly improved the replacement therapy of hemophilia A with a positive impact on the health status of patients, due to the widespread diffusion of prophylaxis to prevent arthropathy, and consequently on their quality of life and life expectancy. In this context, nowadays the most significant debilitating therapeutic complication of hemophilia A, at least in the developed world, has become the development of inhibitors and a continuously increasing number of articles have been published on this topic during the last few years. Inhibitory alloantibodies develop in approximately 20–30% of severe hemophilia A patients, usually within the first 10 exposure days, and render ineffective FVIII substitutive treatment precluding the access of hemophiliacs to a safe and effective standard of care and predisposing to an unacceptably high risk of morbidity and mortality.Citation1,Citation2

The research in this area has been mainly directed at the identification of possible risk factors contributing to inhibitor development, and previously untreated patients (PUPs) have been considered the ideal population for studying the etiology of this phenomenon.Citation3 These studies have revealed the importance of genetic (e.g. ethnicity, FVIII gene mutations (large deletions, nonsense mutations), major histocompatibility complex (MHC) genotype, polymorphisms of immune-responses genes (interleukin-10, tumor necrosis factor-a, cytotoxic T-lymphocyte antigen-4)) and environmental (e.g. the number of FVIII exposure days, age at first exposure of FVIII concentrate, type of FVIII concentrate administered, and modality of treatment) risk factors in the development of inhibitors.Citation4Citation6 All these lines of evidence document that inhibitor formation in hemophilia is a complex multifactorial process.

In this narrative review, I will focus on the role of non-genetic factors in determining the risk of inhibitor development in PUPs with severe hemophilia A and, in particular, I will analyze the published literature data on three of them, which are actually still controversial and debated: age and intensity at first treatment, prophylaxis, and type of FVIII product.

Age and intensity at first FVIII exposure

During the last few years, there has been growing interest in the identification of potentially modifiable factors predisposing the patient to inhibitor development.Citation7 The role of these non-genetic factors has been increasingly supported by evolving concepts of the immune response based on the ‘danger model’Citation8: the immune system is activated by alarm signals arising from the injured tissues to a greater extent than by the recognition of non-self. In this respect, the presentation of the exogenous FVIII may not be sufficient for initiating an immune response. In the presence of danger conditions (i.e. severe bleeds, trauma, or surgery with major tissue injury), the foreign protein is intensively presented (high-dose and/or prolonged treatment) in association with signals that up-regulate the cellular T- and B-lymphocyte response.

This hypothesis is supported by a number of studies showing a key role of the intensity and age of treatment at the first FVIII exposure ().Citation9Citation16 For instance, in a Spanish cohort of 62 children with severe hemophilia A, 24% of whom developed persistent inhibitors, a statistically significant difference (P = 0.03) in the cumulative incidence of laboratory-detected inhibitor formation based on age at first exposure was demonstrated.Citation9 A cohort study from the Netherlands of 81 children with severe hemophilia A confirmed this trend, assessing the incidence at 100 exposure days.Citation10 An Italian case–control study of children treated exclusively with recombinant FVIII (rFVIII) compared 60 children with inhibitors to 48 inhibitor-free controls. Although there seemed to be an increased trend for inhibitor formation if patients were treated at an early age, this did not hold true after adjustment for genetic factors.Citation11 In a retrospective French cohort study, children treated for the first time after 12 months of age showed, at a multivariate analysis, 0.3-fold fewer inhibitors than those treated before 6 months of age.Citation12 In another retrospective cohort study from the United Kingdom of 348 children with severe hemophilia A, 20% developed an inhibitor and 10% of these were high titer. When correlating this to age at first exposure, there was a significant decrease in inhibitor development when children were treated after they were 18 months old (20–26% at ages 0–18 months and 9% at age >18 months).Citation13 In the retrospective CANAL (Concerted Action on Neutralizing Antibodies in Severe Hemophilia A) study, which investigated 366 consecutive PUPs born between 1990 and 2000 from 14 centers in Europe and Canada,Citation14 and in the combined analysis of data on 236 patients from the four rFVIII registration PUP studies,Citation15 surgical and prolonged (≥5 days) FVIII exposure at first treatment, and high FVIII dose (≥50 IU/kg) during the first 50 exposure days were associated with a two-fold to three-fold increase in the risk of inhibitor development. In addition, an association between age at the first treatment (<1 month versus >18 months) and inhibitor development was found at univariate but not at multivariariate analyses.Citation14 In a recent retrospective case–control study, only high-intensity treatments (treatment peaks of 5 exposure days or more and high-frequency treatment) were found to be an independent risk factor for inhibitor development.Citation16 Accordingly, an intensive treatment at initial exposure has been considered the most significant determinant (three points) in the prognostic score, which combined genetic- and treatment-related risk factors, determined from the CANAL data.Citation17 Finally, a recent systematic review by Eckhardt et al.Citation18 on surgery and inhibitor development in hemophilia A patients found that intensive FVIII treatment for surgery at first exposure leads to a higher inhibitor risk compared with intensive treatment for bleeding or prophylaxis (OR 4.1; 95% CI 2.6–6.5).

Table 1. Age and intensity at first FVIII exposure and inhibitor development in severe hemophilia A patients: analysis of the most important published studies

On the other hand, it is likely that the conflicting data from the literature on this issue could be attributable to the presence of several genetic and non-genetic confounders, such as FVIII mutation, product type, ethnicity, family history, etc., which further testify the complex multifactorial nature of inhibitor formation.

Prophylaxis

According to the danger model, a number of studies have assessed whether regular exposure to lower doses of antigen in the absence of danger signals, which occurs during prophylaxis, may induce the tolerization of the foreign protein.

After some indirect data or non-controlled studies,Citation19,Citation20 a protective effect of prophylaxis against the development of inhibitors was shown at multivariate analysis in a case–control Italian study,Citation11 which reported a 70% reduction of inhibitor risk in children starting prophylaxis at a median age of 35 months, compared with children receiving on-demand treatment. Similar findings came from the large multinational CANAL study, in which early regular prophylaxis (introduced at a median age of 20 months) was an independent predictor associated with a 60% reduction of risk of inhibitor development as compared with on-demand treatment.Citation14

Starting from these pathophysiological and clinical observations, Kurnik et al.Citation21 demonstrated that the incidence of inhibitors was markedly reduced by early prophylaxis with once-weekly low-dose FVIII during the first 20 exposure days, thereby minimizing danger signals related to the need of intensive exposure to FVIII. Interestingly, only one of 26 patients receiving such a new regimen developed inhibitors compared to 14 of 30 children retrospectively analyzed treated with standard prophylaxis, resulting in a >90% reduction of risk. However, preliminary results from 606 patients enrolled in the RODIN (Research Of Determinants of INhibitor development) study, recently published in abstract form showed that regular prophylaxis prevented the development of late inhibitors (after about 20 exposure days) for both all the inhibitors and high responder inhibitors.Citation22

Type of FVIII product

The role of FVIII replacement therapy (i.e. plasma-derived (pd) versus recombinant products) in the likelihood of developing inhibitors has been explored by a number of observational multicenter PUP studies during the last two decades and some investigators have postulated that pdFVIII concentrates might have a lower inhibitor incidence thanks to the protective effect of their von Willebrand factor (VWF), which would mask the epitope sites of inhibitors on FVIII molecule or would protect against FVIII by dendritic cells, thus preventing presentation to CD4+ cells and subsequent antibody production.Citation23 However, several criticisms can be raised against those in vitro studies supporting the first mechanism due to the difficulty in reproducing in the laboratory the physiological in vivo conditions (where circulating VWF is in great excess compared to FVIII and immediately binds exogenous FVIII molecules).Citation24 Furthermore, the fact that VWF attached to FVIII masks the C2 domain on the FVIII molecule, and are identified to be significant in antibody production, does not account for alloantibodies directed to the A2 or A3 domains.Citation25 Contradictory studies have recently been published on the second mechanism.Citation26,Citation27

Clinical studies have also yielded conflicting results.Citation23 A prospective study was conducted by Mauser-Bunschoten et al.Citation28 that compared the incidence of inhibitors in 59 severe hemophilia A PUPs, who were initially treated with cryoprecipitate or intermediate purified FVIII concentrates, with that in 22 patients exclusively treated with monoclonally purified FVIII and rFVIII products. The inhibitor incidence rate was very similar between the two groups (24 versus 23%, respectively). In another prospective, comparative study of PUPs with moderate to severe hemophilia A, Kreuz et al.Citation29 detected inhibitors in 18/51(35%) patients treated with pdFVIII and 4/21(19%) patients treated with rFVIII. In the severe subgroup, inhibitors developed in 46% of pdFVIII- and 36% of rFVIII-treated patients. No difference was observed in the development of high-titer inhibitors among the severe group treated with pdFVIII (13/35; 37%) compared with rFVIII (4/11; 36%). In the prospective German Thrombosis and Haemostasis-Previously untreated patients (GTH-PUP) study, a preliminary evaluation in 2003 demonstrated a non-statistically significant difference in the cumulative inhibitor incidence in 57 severe hemophilia A patients treated with one pdFVIII concentrate and 47 patients using one rFVIII product (21 versus 36%, P = 0.08).Citation30

Goudemand et al.Citation12 reported a more than doubled risk of inhibitors in a historical cohort of 86 severe hemophilia A PUP patients treated with a full-length rFVIII product (inhibitor risk, 31%) relative to a similar cohort of 62 patients treated with a single pdFVIII product containing large amounts of VWF (inhibitor risk 11%; adjusted relative risk (RR) 2.4; 95% CI 1.0–5.8; P = 0.049). When only high-titer inhibitors were considered at multivariate analysis, the statistical evidence disappeared (cumulative inhibitor incidence; 5% with pdFVIII versus 15% with rFVIII, P = 0.157). Similarly, in a retrospective cohort study on 348 severe hemophilia A children, Chalmers et al.Citation13 observed a higher risk of inhibitors in patients initially treated with rFVIII products than in those treated with pdFVIII concentrates (27 versus 14%, respectively; P = 0.009); however, this difference was no longer statistically significant when high-titer inhibitors were considered (15 versus 10%, P = 0.173) or at multivariate analysis including other potential risk factors (OR 1.83, 95% CI 0.9–3.72). In the CANAL study,Citation31 the risk of inhibitors was not lower in the patients receiving pdFVIII compared with those treated with rFVIII (RR for all inhibitors, 0.8; 95% CI 0.5–1.3; P = 0.34; RR for high-titer inhibitors, 0.9; 95% CI 0.8–2.5; P = 0.72) and no difference was detectable when low- or high-VWF content products were separately considered. Furthermore, the switching between FVIII products did not increase the risk of inhibitors (RR 1.1; 95% CI 0.6–1.8).

In 2003, Wight and PaisleyCitation32 published a systematic review investigating the association of FVIII product type with inhibitor formation. Fifty relevant retrospective or prospective studies were identified in the literature. A comparison of 13 studies on PUPs found that patients treated with pdFVIII had a lower cumulative inhibitor incidence than those treated with rFVIII. In patients treated with pdFVIII, the cumulative inhibitor incidence ranged from 0 to 12.4% (weighted mean: 6.8%) for all inhibitors and from 0 to 2.5% (weighted mean: 1.4%) for high responders (>5 Bethesda Units, BU). In contrast, patients treated with rFVIII reported cumulative inhibitor rates between 36 and 38.7% (weighted mean: 37.5%) for all patients; for high responders, the inhibitor incidence ranged from 11.3 to 18% (weighted mean: 15.1%). However, several methodological criticisms have been raised against this review which compared very heterogeneous trials in terms of study design (e.g. prospective/retrospective, the frequency and method of inhibitor testing, length of follow-up) and study populations (ethnicity, type of gene mutation, definition of disease severity, age at first exposure to FVIII, the number of exposure days, treatment regimen, etc.),Citation33 making it impossible to draw any conclusion based on the comparison of inhibitor incidence of the different products across studies.

The more recent meta-analysis by Iorio et al.Citation34 identified 2094 patients, from 24 retrospective and prospective studies, developing 420 inhibitors. The pooled incidence inhibitor rate was 14.3% for pdFVIII concentrates and 27.4% for rFVIII products (P < 0.001), which however lost statistical significance at multivariate analysis, where the study design, study period, testing frequency, and median follow-up were found to explain most of the variability. Similar results were observed when the analysis was restricted to the 19 prospective studies (9.1% for pdFVIII concentrates and 23.7% for rFVIII products, P < 0.001). However, the statistical significance disappeared again when only high-titer inhibitors were considered (6.0% with pdFVIII versus 19.4% with rFVIII products, P = 0.195).

In an attempt to control as much as possible the variables of the different studies, to render the data more homogeneous, we have conducted a systematic review and meta-analysis using selective criteria (i.e. only prospective studies, PUPs, and severe hemophilia A patients).Citation35 The study included data from 800 PUPs with severe hemophilia A enrolled in 25 prospective studies and found no statistically significant difference in inhibitor rate between patients treated with plasma-derived and recombinant FVIII products (21 versus 27%). Similarly, high-titer inhibitors did not differ significantly between the two groups (14% with plasma-derived versus 16% with recombinant FVIII products). summarizes the results of the most important comparative studies and of the three published systematic reviews. Interestingly, comparing the results of our meta-analysisCitation35 with those of Iorio et al.Citation34, it emerges that while the rate of inhibitor (overall and high-responding) development with rFVIII products is very similar between the two studies, the difference becomes substantial when pdFVIII products are analyzed. This is due, in our opinion, to the lower quality of the pdFVIII studies as compared with those more recent evaluating rFVIII products. This was clearly outlined during the quality assessment (using the Newcastle-Ottawa Scale (NOS) and Strengthening the Reporting of Observational studies in Epidemiology (STROBE)) and with an ad hoc score system (, unpublished data), which evaluated seven items (population size, median follow-up, exposure days, definition of severity of hemophilia, inhibitor lower limit, pharmacokinetic study, inhibitor screening frequency). In particular, the latter technical quality score, which was created in order to evaluate some specific characteristics not considered in NOS and STROBE checklists, documented a statistically significant superiority of studies on rFVIII products as compared with studies on pdFVIII concentrates (median values 9 versus 14; P = 0.01).Citation36Citation58 This difference, probably due to the fact that rFVIII studies were conducted more recently and thus used better designed protocols than pdFVIII studies, could explain the lower inhibitor incidence, although not statistically significant, found in the latter studies.

Table 2. Summary of the main results of the published comparative studies and systematic reviews comparing the inhibitor development with the use of rFVIII or pdFVIII concentrates in PUPs with severe hemophilia A

Table 3. Ad hoc quality score of the studies included in the meta-analysis by Franchini et al.Citation35 (unpublished data)

Conclusions

The analysis of the current literature data further confirms the complex multifactorial nature of inhibitor development in severe hemophilia A. However, while genetic inhibitor determinants have been well characterized in studies and meta-analysis, less uncertainty still exists as regards environmental factors. Indeed, there is some concern that factor VIII exposure at a younger age, especially less than 6 months of age, contributes to alloantibody formation, although the data are quite conflicting. More convincing data have been published on the role of intensity of treatment (i.e. high dose and/or prolonged treatment), especially in the presence of danger conditions such as surgery, and inhibitor development.

The published literature data suggest a possible protective role for prophylaxis against inhibitor development, especially for late inhibitors (≥20 exposure days).

Finally, there is no conclusive evidence that pdFVIII and rFVIII products are associated with different inhibitor risks. To further address these issues, a number of prospective studies and registries have been started, such as SIPPET, PEDNET, RODIN, GTH-PUP Study and EUHASS. The completion of them will probably help us to answer this issue. For instance, a preliminary 2-year analysis of the data from the prospective registry EUHASS (European Haemophilia Safety Surveillance System) found almost the same inhibitor incidence with pdFVIII and rFVIII products (27 versus 25%).

In conclusion, the studies on the environmental risk factors for inhibitor development in PUPs are very important because while they contribute to elucidate the complex pathophysiology of inhibitor formation, on the other hand they are modifiable and thus represent the ideal target for prevention strategies with the aim of modifying the natural history of alloantibodies, especially in patients with high-risk molecular defects.

References

  • Coppola C, Santoro C, Tagliaferri A, Franchini M, Di Minno G. Understanding inhibitor development. Haemophilia. 2010;16 (Suppl 1):13–9.
  • Franchini M, Coppola A, Molinari AC, Santoro C, Schinco P, Speciale V, et al. Forum on: the role of recombinant factor VIII in children with severe haemophilia A. Haemophilia. 2009;15:578–86.
  • Gomperts ED. The need for previously untreated patient population studies in understanding the development of factor VIII inhibitors. Haemophilia. 2006;12:573–8.
  • Lee CA, Lillicrap D, Astermark J. Inhibitor development in hemophiliacs: the roles of genetic versus environmental factors. Semin Thromb Haemost. 2006;32 (Suppl. 2):10–14.
  • Cox-Gill J. The role of genetics in inhibitor formation. Thromb Haemost. 1999;82:500–4.
  • Astermark J. Overview of inhibitors. Semin Hematol. 2006;43 (2 Suppl.):S3–S7.
  • Gouw SC, van den Berg HM. The multifactorial etiology of inhibitor development in hemophilia: genetics and environment. Semin Thromb Hemost. 2009;35:723–34.
  • Matzinger P. The danger model: a renewed sense of self. Science. 2002;8:76–82.
  • Lorenzo JI, Lopez A, Altisent C, Aznar JA. Incidence of factor VIII inhibitors in severe haemophilia: the importance of patient age. Br J Haematol. 2001;113:600–3.
  • van der Bom JG, Mauser-Bunschoten P, Fischer K, van den Berg HM. Age at first treatment and immune tolerance to factor VIII in severe hemophilia. Thromb Haemost. 2003;89:475–9.
  • Santagostino E, Mancuso ME, Rocino A, Mancuso G, Mazzucconi MG, Tagliaferri A, et al. Environmental risk factors for inhibitor development in children with haemophilia A: a case–control study. Br J Haematol. 2005;130:422–7.
  • Goudemand J, Rothschild C, Demiguel V, Vinciguerrat C, Lambert T, Chambost H, et al. Influence of the type of factor VIII concentrate on the incidence of factor VIII inhibitors in previously untreated patients with severe hemophilia A. Blood. 2006;107:46–51.
  • Chalmers EA, Brown SA, Keeling D, Liesner R, Richards M, Stirling D, et al. Early factor VIII exposure and subsequent inhibitor development in children with severe haemophilia A. Haemophilia. 2007;13:149–55.
  • Gouw SC, van der Bom JG, van den Berg HM. Treatment-related risk factors of inhibitor development in previously untreated patients with hemophilia A: the CANAL cohort study. Blood. 2007;109:4648–54.
  • Gouw SC, van den Berg HM, le Cessie S, van der Bom JG. Treatment characteristics and the risk of inhibitor development: a multicenter cohort study among previously untreated patients with severe hemophilia A. J Thromb Haemost. 2007;5:1383–90.
  • Maclean PS, Richards M, Williams M, Collins P, Liesner R, Keeling DM, et al. Paediatric Working Party of UKHCDO. Treatment related factors and inhibitor development in children with severe haemophilia A. Haemophilia. 2011;17:282–7.
  • Ter Avest PC, Fisher K, Mancuso ME, Santagostino E, Yuste VJ, van den Berg HM, et al. CANAL Study Group. Risk stratification for inhibitor development at first treatment for severe hemophilia A: a tool for clinical practice. J Thromb Haemost. 2008;6:2048–54.
  • Eckhardt CL, van der Bom JG, van der Naald M, Peters M, Kamphuisen PW, Fijnvandraat K. Surgery and inhibitor development in hemophilia A: a systematic review. J Thromb Haemost. 2011;9:1948–58.
  • Yee TT, Beeton K, Griffioen A, Harrington C, Miners A, Lee CA, et al. Experience of prophylaxis treatment in children with severe haemophilia. Haemophilia. 2002;8:76–82.
  • Morado M, Villar A, Jiménez Yuste V, Quintana M, Hernandez Navarro F. Prophylactic treatment effects on inhibitor risk: experience in one centre. Haemophilia. 2005;11:79–83.
  • Kurnik K, Bidlingmaier C, Engl W, Chehadeh H, Reipert B, Auerswald G. New early prophylaxis regimen that avoids immunological danger signals can reduce FVIII inhibitor development. Haemophilia. 2010;16:256–62.
  • Gouw SC, van den Berg M, van der Bom J. Factor VIII prophylaxis and inhibitor development in previously untreated patients with severe hemophilia A: the RODIN study. Haemophilia. 2012;18(Suppl. 3):89, (abstract).
  • Franchini M, Lippi G. VWF-containing FVIII concentrates and inhibitors in hemophilia a: a critical literature review. Thromb Haemost. 2010;104:931–40.
  • Verbruggen B, van Heerde W, Budde U. Methodological shortcomings in assessment of factor VIII concentrate inhibition. Haemophilia. 2007;13:680–1.
  • Suzuki T, Arai M, Amano K, Kagawa K, Fukutake K. Factor VIII inhibitor antibodies with C2 domain specificity are less inhibitory to factor VIII complexed with von Willebrand factor. Thromb Haemost. 1996;76:749–54.
  • Dasgupta S, Repesse Y, Bayry J, Navarrete AM, Wootla B, Delignat S, et al. VWF protects FVIII from endocytosis by dendritic cells and subsequent presentation to immune effectors. Blood. 2007;109:610–2.
  • Pfistershammer K, Stockl J, Siekmann J, Turecek PL, Schwarz HP, Reipert BM. Recombinant factor VIII and factor VIII–von Willebrand factor complex do not present danger signals for human dendritic cells. Thromb Haemost. 2006;96:309–16.
  • Mauser-Bunschoten EP, van der Bom JG, Bongers M, Twijnstra M, Roosendaal G, Fischer K, et al. Purity of factor VIII product and incidence of inhibitors in previously untreated patients with haemophilia A. Haemophilia. 2001;7:364–8.
  • Kreuz W, Ettingshausen CE, Zyschka A, Oldenburg J, Saguer IM, Ehrenforth S, et al. Inhibitor development in previously untreated patients with hemophilia A: a prospective long-term follow-up comparing plasma-derived and recombinant products. Semin Thromb Hemost. 2002;28:285–90.
  • Kreuz W, Ettingshausen CE, Auerswald G, Saguer IM, Becker S, Funk M, et al. Epidemiology of inhibitors and current treatment strategies. Haematologica. 2003;88:pEREP04.
  • Gouw SC, van der Bom JG, Auerswald G, Ettinghausen CE, Tedgård U, van den Berg HM. Recombinant versus plasma-derived factor VIII products and the development of inhibitors in previously untreated patients with severe hemophilia A: the CANAL cohort study. Blood. 2007;109:4693–7.
  • Wight J, Paisley S. The epidemiology of inhibitors in haemophilia A: a systematic review. Haemophilia. 2003;9:418–35.
  • Scharrer I, Ehrlich HJ. Reported inhibitor incidence in FVIII PUP studies: comparing apples with oranges? Haemophilia. 2004;10:197–8.
  • Iorio A, Halimeh S, Holzhauer S, Goldenberg N, Marchesini E, Marcucci M, et al. Rate of inhibitor development in previously-untreated hemophilia a patients treated with plasma-derived or recombinant factor VIII concentrates. A systematic review. J Thromb Haemost. 2010;8:1256–65.
  • Franchini M, Tagliaferri A, Mengoli C, Cruciani M. Cumulative inhibitor incidence in previously untreated patients with severe hemophilia A treated with plasma-derived versus recombinant factor VIII concentrates: a critical systematic review. Crit Rev Oncol Hematol. 2012;81:82–93.
  • Lusher JM, Salzman PM. Viral safety and inhibitor development associated with factor VIIIC ultra-purified from plasma in haemophiliacs previously unexposed to factor VIII C concentrates. Semin Hematol. 1990;27 (Suppl. 2):1–7.
  • Lusher JM. Viral safety and inhibitor development associated with monoclonal antibody-purified FVIII:C. Ann Hematol. 1991;63:138–41.
  • Ehrenforth S, Kreuz W, Scharrer I, Linde R, Funk M, Güngör T, et al. Incidence of development of factor VIII and factor IX inhibitors in haemophiliacs. Lancet. 1992;339:594–8.
  • Ljung R, Petrini P, Lindgren A, Tengborn L, Nilsson IM. Factor VIII and factor IX inhibitors in haemophiliacs. Lancet. 1992;339:1550.
  • Addiego JE-J, Gomperts E, Liu SL, Bailey P, Courter SG, Lee ML, et al. Treatment of hemophilia A with a highly purified factor VIII concentrate prepared by anti-FVIII:C immunoaffinity chromatography. Thromb Haemost. 1992;67:19–27.
  • de Biasi R, Rocino A, Papa E, Salerno E, Mastrullo L, De Blasi D. Incidence of factor VIII inhibitor development in hemophilia A patients treated with less pure plasma derived concentrates. Thromb Haemost. 1994;71:544–7.
  • Yee TT, Williams MD, Hill FG, Lee CA, Pasi KJ. Absence of inhibitors in previously untreated patients with severe haemophilia A after exposure to a single intermediate purity factor VIII product. Thromb Haemost. 1997;78:1027–9.
  • El Alfy MS, Tantawy AA, Ahmed MH, Abdin IA. Frequency of inhibitor development in severe haemophilia A children treated with cryoprecipitate and low-dose immune tolerance induction. Haemophilia. 2000;6:635–8.
  • Bray GL, Gomperts ED, Courter SG. A multicenter study of recombinant factor VIII (recombinate): safety, efficacy, and inhibitor risk in previously untreated patients. Blood. 1997;83:2428–35.
  • Gruppo R, Chen H, Schroth P, Bray GL. Safety and immunogenicity of recombinant factor VIII (Recombinate) in previously untreated patients: a 7.3 year update. Haemophilia. 1998;4:228, ( abstract no. 291).
  • Goodeve AC, Williams I, Bray GL, Peake IR. Relationship between factor VIII mutation type and inhibitor development in a cohort of previously untreated patients treated with recombinant factor VIII (Recombinate). Recombinate PUP Study Group. Thromb Haemost. 2000;83:844–8.
  • Rothschild C, Laurian Y, Satre EP, Borel Derlon A, Chambost H, Moreau P, et al. French previously untreated patients with severe hemophilia A after exposure to recombinant factor VIII: incidence of inhibitor and evaluation of immune tolerance. Thromb Haemost. 1998;80:779–83.
  • Lusher JM, Arkin S, Abildgaard CF, Schwartz RS. Recombinant factor VIII for the treatment of previously untreated patients with haemophilia A. Safety, efficacy and development of inhibitors. Kogenate previously untreated patient study group. N Engl J Med. 1993;328:453–9.
  • Lusher J, Abildgaard C, Arkin S, Mannucci PM, Zimmermann R, Schwartz L, et al. Human recombinant DNA-derived antihemophilic factor in the treatment of previously untreated patients with hemophilia A: final report on a hallmark clinical investigation. J Thromb Haemost. 2004;2:574–83.
  • Yoshioka A, Fukutake K, Takamatsu J, Shirahata A, Kogenate Post-Marketing Surveillance Study Group. Evaluation of a recombinant factor VIII preparation (Kogenate) in previously untreated patients with hemophilia A. Int J Hematol. 2003;78:467–74.
  • Giangrande PLF, for the Kogenate Bayer Study Group. Safety and efficacy of Kogenate Bayer in previously untreated patients (PUPs) and minimally treated patients (MTPs). Haemophilia. 2002;8 (suppl. 2):19–22.
  • Kreuz W, Gill JC, Rothschild C, Manco-Johnson MJ, Lusher JM, Kellermann E, et al. Full-length sucrose-formulated recombinant factor VIII for treatment of previously untreated or minimally treated young children with severe haemophilia A: results of an international clinical investigation. Thromb Haemost. 2005;3:457–67.
  • Oldenburg J, Ivaskevicius V, Schröder J, Müller CR, Ganguly A. Genetic background and inhibitors in previously untreated or minimally treated young patients with severe haemophilia A treated with sucrose-formulated recombinant factor VIII. Thromb Haemost. 2006;95:903–5.
  • Musso R, Santagostino E, Faradji A, et al., Kogenate Bayer European PMS Study Group. Safety and efficacy of sucrose-formulated full-length recombinant factor VIII: experience in the standard clinical setting. Thromb Haemost. 2008;99:52–8.
  • Lusher JM, Lee CA, Kessler CM, Bedrosian CL. The safety and efficacy of B-domain deleted recombinant factor VIII concentrate in patients with severe haemophilia A. Haemophilia. 2003;9:38–49.
  • Lusher JM, Roth DA. The safety and efficacy of B-domain deleted recombinant factor VIII concentrates in patients with severe haemophilia: an update. Haemophilia. 2005;11:292–3.
  • Courter SG, Bedrosian CL. Clinical evaluation of B-domain deleted recombinant factor VIII in previously untreated patients. Semin Hematol. 2001;38:52–9.
  • Pollmann H, Externest D, Ganser A, Eifrig B, Kreuz W, Lenk H, et al. Efficacy, safety and tolerability of recombinant factor VIII (REFACTO) in patients with haemophilia A: interim data from a postmarketing surveillance study in Germany and Austria. Haemophilia. 2007;13:131–43.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.