Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters

Publication year range
1.
Br J Haematol ; 178(1): 20-31, 2017 07.
Article in English | MEDLINE | ID: mdl-28387451

ABSTRACT

Minimizing the risk of inhibitor development by acting on modifiable risk factors remains a sensible goal for treatment optimization in haemophilia A. By critically appraising published studies assessing inhibitor development, this review addresses the role of studies in previously untreated patients (PUPs) for establishing the immunogenicity of new concentrates, suggest novel research design to be adopted in future studies and discuss clinical practice implications of the reported differential immunogenicity of Kogenate Bayer and Advate factor VIII concentrates. Three considerations are relevant here: (i) all of the existing concentrates, when tested following the International Society on Thrombosis and Haemostasis Scientific and Standardization Committee recommendation, were shown to be safe; as a consequence, (ii) when considering using any newly introduced product, one should be aware that it could, in future, turn out to be as immunogenic as Kogenate Bayer, and (iii) at the population level, it might be wiser not to use Kogenate Bayer in PUPs, if the choice is against Advate. When presenting the risk of developing inhibitors to the individual patient (or their family), the message remains that the risk can be as high as 40%, without any efficient instrument to predict individual inhibitor risk. Patients should be invited to enrol into a randomized registry trial, including random assignment to trials with new investigational products.


Subject(s)
Coagulants/therapeutic use , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Isoantibodies/blood , Blood Coagulation Factor Inhibitors/blood , Coagulants/immunology , Factor VIII/immunology , Hemophilia A/immunology , Humans
2.
Lancet ; 388(10040): 187-97, 2016 Jul 09.
Article in English | MEDLINE | ID: mdl-26897598

ABSTRACT

Haemophilia A and B are hereditary haemorrhagic disorders characterised by deficiency or dysfunction of coagulation protein factors VIII and IX, respectively. Recurrent joint and muscle bleeds lead to severe and progressive musculoskeletal damage. Existing treatment relies on replacement therapy with clotting factors, either at the time of bleeding (ie, on demand) or as part of a prophylactic schedule. The major complication of such therapy is the development of neutralising antibodies (ie, inhibitors), which is most frequent in haemophilia A. Treatment might improve considerably with the availability of new modified drugs, which might overcome existing prophylaxis limitations by reducing dosing frequency and thereby rendering therapy less distressing for the patient. Subcutaneous administration of some new therapies would also simplify prophylaxis in children with poor venous access. Gene therapy has the potential for a definitive cure, and important results have been obtained in haemophilia B. Despite improvements in haemophilia care, the availability of clotting factor concentrates for all affected individuals worldwide remains the biggest challenge.


Subject(s)
Coagulants/therapeutic use , Factor IX/therapeutic use , Factor VIII/therapeutic use , Hemophilia A/therapy , Hemophilia B/therapy , Hemorrhage/therapy , Antibodies, Neutralizing/immunology , Coagulants/immunology , Early Medical Intervention , Factor IX/immunology , Factor VIII/immunology , Genetic Therapy , Hemophilia A/diagnosis , Hemophilia A/immunology , Hemophilia B/diagnosis , Hemophilia B/immunology , Hemorrhage/prevention & control , Humans
3.
Blood Cells Mol Dis ; 66: 1-5, 2017 07.
Article in English | MEDLINE | ID: mdl-28689155

ABSTRACT

BACKGROUND: Therapy application and monitoring of patients with hemophilia A (HA) and inhibitors are challenging. In the current study, combined FVIII - bypass therapy was implemented for a cohort of severe HA patients with inhibitors. METHODS: Plasma of 15 HA patients with inhibitors was spiked ex vivo with FVIII, rFVIIa, FEIBA and their combinations and thrombin generation (TG) was studied. Some patients who experienced hemarthroses or required minor surgeries were treated by a combined concomitant administration of FVIII+FEIBA as IV bolus doses. RESULTS: TG spiking studies showed individual responses not correlated to inhibitor titer. Combinations of agents augmented TG as compared to any single agent, while combined FVIII+FEIBA yielded the highest TG, supporting it as a potential treatment. Following emergent successful surgery of child treated by concomitant FVIII+FEIBA, a total of 396 episodes in 7/15 patients were treated with concomitant FVIII+FEIBA. Five patients were treated for bleeding episodes only, whereas 2 were children undergoing immune tolerance induction (ITI) with FEIBA prophylaxis. Four minor surgeries were performed on FVIII+FEIBA repeated infusions. Neither thrombosis nor any other adverse events were documented. CONCLUSION: A combination of FVIII+FEIBA may be effective and safe as an alternative treatment option for some high-responding inhibitor patients.


Subject(s)
Drug Therapy, Combination/methods , Hemophilia A/drug therapy , Hemostatics/therapeutic use , Adolescent , Adult , Antibodies/analysis , Blood Coagulation Factors/therapeutic use , Child , Coagulants/immunology , Coagulants/therapeutic use , Factor VIII/immunology , Factor VIII/therapeutic use , Hemophilia A/immunology , Humans , Treatment Outcome , Young Adult
4.
Haemophilia ; 22(3): 334-41, 2016 May.
Article in English | MEDLINE | ID: mdl-26843214

ABSTRACT

INTRODUCTION: The occurrence of a neutralizing antibody in previously untreated patients (PUPs) with haemophilia A appears to be the result of an intricate interplay of both genetic and environmental factors. Recently, the type of factor VIII (FVIII) product used in the PUPs population has been implicated as a risk factor for inhibitor development. AIM: The aim of this review was to explore in a systematic manner potential hypotheses for the product-related findings in these studies (i.e. differences in the expression system of the cell lines used to produce recombinant FVIII [rFVIII], differences in the administered antigen load or changes in clinical practice over time). RESULTS: Review of the available clinical studies illustrates the high degree of variability for the risk of inhibitor development for the same products across different studies. Differences in cell lines or antigen load were not found to provide a reasonable explanation. CONCLUSION: The possibility of changes in clinical practice over time and patient selection bias (i.e. the preferential use of one product over another in patients at higher risk for inhibitors) offers a potential explanation and should be carefully considered when evaluating the studies.


Subject(s)
Antibodies, Neutralizing/blood , Coagulants/therapeutic use , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Coagulants/immunology , Factor VIII/immunology , Humans , Protein Processing, Post-Translational , Risk Factors , von Willebrand Factor/chemistry , von Willebrand Factor/metabolism
5.
Haemophilia ; 22(5): 772-9, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27439658

ABSTRACT

INTRODUCTION: The most serious complication in haemophilia A (HA) replacement therapy with coagulation factor VIII (FVIII) is neutralizing antibodies, i.e. inhibitors. It has been hypothesized that danger signals generated during a bleed might have an adjuvant effect on the immune response to FVIII in on-demand treatment, increasing the inhibitor risk. AIM: To compare the antibody response to treatment with recombinant human FVIII (rhFVIII) in relation to induced knee joint bleeds and treatment without concurrent bleeds in a HA rat model. METHOD: HA rats were divided into two groups: one group (n = 10) receiving three needle induced knee joint bleeds 14 days apart and a control group (n = 9) receiving three sham procedures. Three hours after each injury/sham 50 IU kg(-1) rhFVIII was administrated intravenously. Subsequently, both groups continued rhFVIII treatment for another 9 weeks. Binding antibodies were analysed using an enzyme-linked immunosorbent assay and neutralizing antibodies using a Bethesda-like assay. RESULTS: Rats in the knee-bleed group developed a significantly faster inhibitor response and reached significantly higher inhibitor levels. In the knee-bleed group, 80% developed inhibitors vs. 33% in the control group, demonstrating a 2.4 times higher inhibitor risk when treating concurrent with bleeds. CONCLUSION: FVIII treatment in relation to a bleed potentiates inhibitor development compared to FVIII treatment alone in this HA rat, indicating that bleeding is a potential danger signal. Our results support the theory that FVIII replacement therapy concurrent with a bleeding episode increases the inhibitor risk, which to the best of our knowledge, has not been confirmed in an animal model before.


Subject(s)
Antibodies, Neutralizing/blood , Autoantibodies/blood , Hemarthrosis/etiology , Hemophilia A/drug therapy , Animals , Coagulants/adverse effects , Coagulants/immunology , Coagulants/therapeutic use , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Factor VIII/adverse effects , Factor VIII/genetics , Factor VIII/immunology , Factor VIII/therapeutic use , Factor VIIa/therapeutic use , Female , Hemarthrosis/prevention & control , Hemophilia A/pathology , Humans , Joints/physiology , Male , Rats , Recombinant Proteins/therapeutic use
6.
Blood ; 121(22): 4449-56, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23502223

ABSTRACT

A major problem in treating hemophilia A patients with therapeutic factor VIII (FVIII) is that 20% to 30% of these patients produce neutralizing anti-FVIII antibodies. These antibodies block (inhibit) the procoagulant function of FVIII and thus are termed "inhibitors." The currently accepted clinical method to attempt to eliminate inhibitors is immune tolerance induction (ITI) via a protocol requiring intensive FVIII treatment until inhibitor titers drop. Although often successful, ITI is extremely costly and is less likely to succeed in patients with high-titer inhibitors. During the past decade, significant progress has been made in clarifying mechanisms of allo- and autoimmune responses to FVIII and in suppression of these responses. Animal model studies are suggesting novel, less costly methods to induce tolerance to FVIII. Complementary studies of anti-FVIII T-cell responses using blood samples from human donors are identifying immunodominant T-cell epitopes in FVIII and possible targets for tolerogenic efforts. Mechanistic experiments using human T-cell clones and lines are providing a clinically relevant counterpoint to the animal model studies. This review highlights recent progress toward the related goals of lowering the incidence of anti-FVIII immune responses and promoting durable, functional immune tolerance to FVIII in patients with an existing inhibitor.


Subject(s)
Factor VIII/immunology , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Hemophilia A/immunology , Immune Tolerance/immunology , Coagulants/immunology , Coagulants/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use
7.
Blood ; 121(17): 3484-92, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23460609

ABSTRACT

Heparin-induced thrombocytopenia (HIT) is an immune-mediated disorder that can cause fatal arterial or venous thrombosis/thromboembolism. Immune complexes consisting of platelet factor 4 (PF4), heparin, and PF4/heparin-reactive antibodies are central to the pathogenesis of HIT. However, the B-cell origin of HIT antibody production is not known. Here, we show that anti-PF4/heparin antibodies are readily generated in wild-type mice on challenge with PF4/heparin complexes, and that antibody production is severely impaired in B-cell-specific Notch2-deficient mice that lack marginal zone (MZ) B cells. As expected, Notch2-deficient mice responded normally to challenge with T-cell-dependent antigen nitrophenyl-chicken γ globulin but not to the T-cell-independent antigen trinitrophenyl-Ficoll. In addition, wild-type, but not Notch2-deficient, B cells plus B-cell-depleted wild-type splenocytes adoptively transferred into B-cell-deficient µMT mice responded to PF4/heparin complex challenge. PF4/heparin-specific antibodies produced by wild-type mice were IgG2b and IgG3 isotypes. An in vitro class-switching assay showed that MZ B cells were capable of producing antibodies of IgG2b and IgG3 isotypes. Lastly, MZ, but not follicular, B cells adoptively transferred into B-cell-deficient µMT mice responded to PF4/heparin complex challenge by producing PF4/heparin-specific antibodies of IgG2b and IgG3 isotypes. Taken together, these data demonstrate that MZ B cells are critical for PF4/heparin-specific antibody production.


Subject(s)
Antibody Formation , Autoantibodies/immunology , B-Lymphocytes/immunology , Heparin/immunology , Platelet Factor 4/immunology , Thrombocytopenia/immunology , Adoptive Transfer , Animals , Anticoagulants/adverse effects , Anticoagulants/immunology , Antigen-Presenting Cells/immunology , Autoantibodies/blood , B-Lymphocytes/chemistry , Coagulants/adverse effects , Coagulants/immunology , Flow Cytometry , Heparin/adverse effects , Immunization , Immunoglobulin Class Switching/immunology , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Factor 4/adverse effects , Receptor, Notch2/physiology , Thrombocytopenia/chemically induced , Thrombocytopenia/diagnosis
8.
Haemophilia ; 21(4): e281-5, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25929310

ABSTRACT

In contrast to haemophilia B, allergic manifestations are rare complications in haemophilia A (HA) patients treated with factor VIII (FVIII) concentrates. Nevertheless, it can be serious and hamper replacement therapy in these cases. The aims of this study were to evaluate the frequency of allergic reaction in a cohort of HA patients treated only with plasma-derived FVIII (pdFVIII) concentrates, and assess the possible immune mechanisms involved. History of allergic reaction was retrospectively assessed. Patients with allergic manifestations were followed, and had plasma samples collected in different timepoints in relation to the allergic episode. These samples were analysed for the presence of inhibitor and anti-FVIII immunoglobulins subclasses. Three of 322 HA patients (0.9%) developed allergic reaction after exposure to pdFVIII products during the last 15 years in our centre. The first patient, with severe HA, without inhibitor, had anti-pdFVIII IgE and IgG4, but no anti-recombinant FVIII (rFVIII) IgE. The second patient, with severe HA, and high-responding inhibitor, presented allergic manifestation with both, pdFVIII concentrate and activated prothrombin complex concentrate. Although anti-pdFVIII and anti-rFVIII IgG4 were detected, no anti-FVIII IgE was present. The third patient, with moderate HA without inhibitor, atopic, had no anti-FVIII immunoglobulin detected, and allergic symptoms disappeared after switching to rFVIII concentrate. This study corroborates the low incidence of allergic reactions in HA patients. In the three cases presented, the anti-FVIII immunoglobulin profile demonstrated that the allergic manifestation was triggered by other proteins contained in pdFVIII products, and not directed to FVIII.


Subject(s)
Factor VIII/therapeutic use , Hemophilia A/drug therapy , Adult , Child, Preschool , Coagulants/adverse effects , Coagulants/immunology , Coagulants/therapeutic use , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Factor VIII/adverse effects , Factor VIII/immunology , Hemophilia A/pathology , Humans , Hypersensitivity/etiology , Hypersensitivity/pathology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Middle Aged , Retrospective Studies , Severity of Illness Index
9.
Haemophilia ; 20 Suppl 6: 2-16, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24975700

ABSTRACT

Development of inhibitory antibodies to infused factor VIII (FVIII) concentrates is the most serious unresolved complication of haemophilia A treatment. Systematic reviews suggest a twofold higher incidence of inhibitors with recombinant (rFVIII) vs. plasma-derived (pdFVIII) FVIII products, but study methodologies vary widely. The lower immunogenicity of pdFVIII concentrates is believed to derive from the presence of von Willebrand factor (VWF) which acts as protector and chaperone for FVIII. Several novel investigations reinforce the protective role of the VWF/FVIII complex in inhibitor development. At the basic science level, numerous in vitro and in vivo experiments have demonstrated that VWF-containing pdFVIII concentrates (pdFVIII/VWF) provide better protection against inhibitor neutralization than rFVIII products. Conformational aspects of the binding between VWF and FVIII are thought to prevent the 'attack' on FVIII by inhibitory antibodies. VWF/FVIII binding is 100% in pdFVIII products but only 80% in recombinant products and this 'free' FVIII may be a target for inhibitory antibodies. At the clinical level, newer strategies to prevent inhibitor development in previously untreated patients with severe haemophilia are under investigation. The concept of early prophylaxis (before the onset of a bleed) is convincing from a theoretical point of view but requires further evaluation. The Study on Inhibitors in Plasma-Product Exposed Toddlers is specifically addressing the issue of relative immunogenicity between classes of FVIII product (recombinant vs. plasma-derived). Currently nearing its target enrolment of 300 patients, this international randomized controlled trial is expected to provide some definitive answers about this ever-present clinical dilemma.


Subject(s)
Coagulants/therapeutic use , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Immune Tolerance/drug effects , von Willebrand Factor/therapeutic use , Blood Coagulation Factor Inhibitors/blood , Coagulants/immunology , Drug Substitution , Factor VIII/antagonists & inhibitors , Factor VIII/immunology , Hemophilia A/immunology , Humans , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , von Willebrand Factor/antagonists & inhibitors , von Willebrand Factor/immunology
10.
Haemophilia ; 20 Suppl 6: 17-26, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24975701

ABSTRACT

The development of alloantibody inhibitors against factor VIII (FVIII) represents the most significant complication of haemophilia care. Inhibitors tend to develop early in the course of treatment in about 20-30% of patients with severe haemophilia who receive on-demand or prophylactic FVIII therapy. Many factors are associated with inhibitor formation, including disease severity, major FVIII gene defects, family history and non-Caucasian race, as well as age at first treatment, intensity of early treatment, use of prophylaxis and product choice. As these latter treatment-related variables are modifiable, they provide opportunity to minimize inhibitor incidence at the clinical level. Data from the Bonn Centre in Germany have indicated an overall success rate of 78% for immune tolerance induction (ITI) therapy, with a failure rate of 15% and with some treatments either ongoing (3%) or withdrawn (4%). Similarly, data from the G-ITI study, the largest international multicentre ITI study using a single plasma-derived (pd) FVIII/von Willebrand factor (VWF) product, have demonstrated success rates (complete and partial) in primary and rescue ITI of 87% and 74%, respectively, with 85% of poor prognosis patients achieving success. Favourable clinical results based on success rates and time to tolerization continue to be reported for use of pdFVIII/VWF in ITI, with pdFVIII/VWF having a particular role in patients who require rescue ITI and those with a poor prognosis for success. Data from prospective, randomized, controlled clinical studies, such as RES.I.ST (Rescue Immune Tolerance Study), are eagerly awaited. Another factor to consider with ITI therapy is cost; preliminary data from an updated decision analytic model have provided early evidence that ITI has an economic advantage compared with on-demand or prophylactic therapy.


Subject(s)
Coagulants/therapeutic use , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Immune Tolerance/drug effects , von Willebrand Factor/therapeutic use , Blood Coagulation Factor Inhibitors/blood , Coagulants/immunology , Cost-Benefit Analysis , Factor VIII/antagonists & inhibitors , Factor VIII/immunology , Hemophilia A/economics , Hemophilia A/immunology , Humans , Immune Tolerance/immunology , von Willebrand Factor/antagonists & inhibitors , von Willebrand Factor/immunology
11.
Haemophilia ; 19 Suppl 1: 2-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23278993

ABSTRACT

Development of alloantibodies against infused factor VIII (FVIII) is the most significant complication of haemophilia care today. Antibodies inactivate the procoagulant activity of FVIII and inhibit patients' response to replacement therapy. As inhibitors tend to develop early in the course of FVIII treatment, the challenge is to bring patients through the critical early phase of FVIII exposure without inhibitor development as the subsequent risk is much lower. Disease severity, major FVIII gene defects, family history and non-Caucasian race are major risk factors for inhibitor development. Other variables thought to play a role in inhibitor formation include age at first treatment, intensity of early treatment, use of prophylaxis and product choice [especially recombinant vs. plasma-derived von Willebrand factor (VWF)-containing concentrates]. As these treatment-related variables are modifiable, they provide opportunity to minimize inhibitor incidence at the clinical level. At present, most data regarding inhibitor development derive from retrospective studies, registry reviews, small case series and uncontrolled studies. Findings have often been conflicting, which precludes drawing definitive conclusions. Nevertheless, some clarity is beginning to emerge. Intensity of early treatment appears to be a stronger risk factor for inhibitor development than timing of first treatment. Controlled early antigen presentation via prophylaxis looks promising, particularly in conjunction with strategies to avoid immunological danger signals, but the timing of introduction and optimal regimen are not yet known. Several reports suggest that plasma-derived VWF-containing FVIII concentrates are less immunogenic than recombinant or VWF-free plasma-derived concentrates, but this is awaiting confirmation in the ongoing prospective Survey of Inhibitors in Plasma-Product Exposed Toddlers study.


Subject(s)
Autoantibodies/blood , Blood Coagulation Factor Inhibitors/blood , Coagulants/therapeutic use , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Coagulants/immunology , Factor VIII/genetics , Factor VIII/immunology , Hemophilia A/immunology , Humans , Immune Tolerance , Polymorphism, Genetic , Risk Factors
12.
Haemophilia ; 19 Suppl 1: 8-11, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23278994

ABSTRACT

Approximately 20-30% of patients with severe haemophilia A develop alloantibodies ('inhibitors') to infused FVIII rendering use of such replacement therapy ineffective. Once an inhibitor emerges, immune tolerance induction (ITI) is the standard treatment. ITI involves giving regular doses of FVIII concentrate to eradicate the inhibitor and achieve immunogenic acceptance of administered FVIII. In the early 2000s, a retrospective analysis of inhibitor patients treated at a single centre in Germany indicated that success rates were higher when patients were treated with von Willebrand factor (VWF)-containing plasma-derived FVIII (pdFVIII/VWF) concentrate compared with recombinant or non-VWF-containing pdFVIII products. Importantly, pdFVIII/VWF as rescue therapy was able to convert 8 of 10 patients who had failed primary ITI with recombinant or non-VWF-containing pdFVIII product. A subsequent study from Italy in patients with poor prognostic factors for ITI success also reported good success rates with pdFVIII/VWF as rescue therapy (53% success; 41% partial success). The Grifols-Immune Tolerance Induction (G-ITI) Study represents the largest group of haemophilia A inhibitor patients treated with a single pdFVIII/VWF concentrate (Alphanate(®)/Fanhdi(®)) to be reported to date. Data have been collected for 95 patients who underwent primary or rescue ITI at 46 centres in Europe and the US. Currently, published data are available for 33 patients in the US cohort (11 centres), and data from the European cohort are being analysed. Both groups contained patients with poor prognostic factors and most patients received a high-dose regimen (≥ 100 IU pdFVIII/VWF kg(-1) daily). As expected, the success rate was better for primary vs. rescue ITI and for patients with good vs. poor prognostic factors. However, more than half the patients in the US cohort receiving rescue ITI achieved success (33% complete success; 20% partial success). These results should encourage clinicians to consider the use of pdFVIII/VWF concentrates for rescue ITI. Published outcomes data from the total global G-ITI cohort (95 patients) are awaited with anticipation.


Subject(s)
Coagulants/immunology , Factor VIII/immunology , Hemophilia A/immunology , Immune Tolerance/drug effects , von Willebrand Factor/immunology , Coagulants/antagonists & inhibitors , Coagulants/therapeutic use , Factor VIII/antagonists & inhibitors , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Humans , von Willebrand Factor/antagonists & inhibitors , von Willebrand Factor/therapeutic use
13.
Haemophilia ; 19 Suppl 1: 12-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23278995

ABSTRACT

Among the proposed predictors for immune tolerance induction (ITI) outcome, the therapeutic regimen - specifically the dose and frequency of administered factor VIII (FVIII) as well as FVIII product type - is intensely debated. Are there any advantages for low-dose regimens (50 IU FVIII kg(-1) three times a week) over high-dose regimens (200 IU FVIII kg day(-1)) or vice versa? Are von Willebrand factor (VWF)-containing plasma-derived concentrates superior to recombinant FVIII concentrates for tolerance induction? A review of the available literature indicates that patients with good prognostic factors can achieve success with either low-dose or high-dose ITI regimens. Retrospective data suggest that patient characteristics such as maximum historical inhibitor titres and pre-ITI inhibitor titres are better predictors of treatment success than dose. Results of the prospective International ITI Study have recently become available. In inhibitor patients with good prognosis, success rates were similar between low-dose (50 IU FVIII kg(-1) three times a week) and high-dose (200 IU FVIII kg(-1) daily) regimens. However, patients receiving low-dose ITI took longer to achieve various ITI milestones and had a significantly higher bleed rate per month compared with the high-dose group (0.62 vs. 0.28; P = 0.00024), findings with important clinical implications. Inhibitor patients with poor prognostic features should be treated with a high-dose protocol. This conclusion is supported by a meta-analysis of the International Immune Tolerance Registry and North American Immune Tolerance Registry and by data from Germany showing good success rates with the high-dose, high-frequency Bonn protocol in poor prognosis patients. Type of concentrate also appears to have an influence on ITI success rates in this patient subgroup, with evidence suggesting an advantage for VWF-containing plasma-derived FVIII concentrates over recombinant or VWF-free concentrates. The ongoing prospective studies REScue Immunotolerance STudy and Observational Immune Tolerance Induction are evaluating ITI outcome with respect to product type and are expected to answer this important clinical question as well as provide greater insight into patient- and therapy-related variables in inhibitor patients with poor prognostic features.


Subject(s)
Coagulants/administration & dosage , Factor VIII/administration & dosage , Hemophilia A/drug therapy , Immune Tolerance/drug effects , Coagulants/antagonists & inhibitors , Coagulants/immunology , Dose-Response Relationship, Immunologic , Factor VIII/antagonists & inhibitors , Factor VIII/immunology , Hemophilia A/immunology , Humans , Prognosis
14.
Haemophilia ; 19 Suppl 1: 18-23, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23278996

ABSTRACT

Immune tolerance induction (ITI) is recognized as the first choice treatment in haemophilic patients with inhibitors, with the aim of restoring safe and effective standard factor VIII replacement and, particularly, prophylaxis in children. For the latter, literature data and clinical practice support the optimal cost utility ratio of ITI. Indeed, the high success rate, the low incidence of inhibitor recurrence after successful ITI and the possibility of preventing joint deterioration, enable one to predict a considerable long-term reduction of costs in the majority of treated patients. Therefore, in spite of high costs and open issues about optimal regimens, ITI is actually attempted in virtually all children with inhibitors. Few patients with long-standing inhibitors presently undergo ITI, particularly in the case of severe bleeding tendency. In this setting, uncertainties concerning management are amplified by the paucity of literature data and psychological reluctance by both patients and treaters due to the perceived poor prognosis and the demanding treatment (also in terms of costs). However, clinical data suggest that the role of age at ITI start and of time interval from inhibitor diagnosis, as predictors of ITI outcome, should be considered in a larger framework of proposed and more established prognostic factors. Moreover, optimising ITI management, particularly with respect to inhibitor titre at ITI start and avoidance of adverse events or interruption of treatment, may also contribute to improve outcomes. Although the economic constraints of the present era significantly affect resources for such a high-cost treatment, the individual cost-utility ratio (bleeding tendency and risk of fatal bleeding, arthropathy and need for orthopaedic surgery, comorbidities, quality of life) should be assessed carefully to determine whether ITI is a suitable option and thus not preclude adults from the opportunity of inhibitor eradication.


Subject(s)
Coagulants/immunology , Factor VIII/immunology , Hemophilia A/drug therapy , Immune Tolerance , Isoantibodies/blood , Coagulants/economics , Coagulants/therapeutic use , Cost-Benefit Analysis , Factor VIII/economics , Factor VIII/therapeutic use , Hemophilia A/economics , Hemophilia A/immunology , Humans
15.
Haemophilia ; 19(2): 281-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23038998

ABSTRACT

Immune tolerance induction (ITI) can eliminate factor VIII (FVIII) inhibitory antibodies that appear during FVIII replacement therapy. If first-line ITI fails, switching to a different FVIII concentrate, especially one containing von Willebrand factor (VWF), has been advocated. The objective of the study was to assess the efficacy and safety of Haemate(®) P, a plasma-derived FVIII concentrate containing high levels of VWF, as ITI in severe haemophilia A patients who had failed at least one prior ITI attempt with a different FVIII concentrate. In this multicentre, observational study, Haemate(®) P was administered at a starting dose of 83-308 IU kg(-1) day(-1) (1500-6000 IU day(-1)). Efficacy was assessed by standard criteria (e.g. Bethesda titre, FVIII recovery and half-life), and bleeding characteristics. Nine patients from six haemophilia centres were treated with Haemate(®) P after failing one (n = 2), two (n = 5) or three (n = 2) prior ITI courses. The median time from inhibitor detection to Haemate(®) P treatment was 5.4 years. The median Haemate(®) P dose was 134 IU kg(-1), and the median treatment duration 32 months. During median of 47 months of follow-up, complete response, partial response and treatment failure were observed in one, three and five patients respectively. Five patients experienced seven adverse events (AEs), including two serious AEs (sepsis). Haemate(®) P was discontinued due to an AE in one patient with a partial response. Haemate(®) P salvage ITI resulted in complete or partial tolerization in four of nine patients (44%) who had failed previous ITI attempts using different FVIII concentrates.


Subject(s)
Coagulants/therapeutic use , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Immune Tolerance/drug effects , von Willebrand Factor/therapeutic use , Adolescent , Adult , Child , Child, Preschool , Coagulants/adverse effects , Coagulants/immunology , Drug Combinations , Factor VIII/adverse effects , Factor VIII/immunology , France , Hemophilia A/immunology , Humans , Male , Middle Aged , Retrospective Studies , Young Adult , von Willebrand Factor/adverse effects , von Willebrand Factor/immunology
16.
Pediatr Nephrol ; 28(5): 823-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23381011

ABSTRACT

BACKGROUND: Nephrotic syndrome (NS) is a recognized complication of immune tolerance induction (ITI) therapy, a treatment strategy used to treat inhibitors in patients with hemophilia B receiving factor IX concentrate. CASE DIAGNOSIS/TREATMENT: We present a 4-year-old boy with hemophilia B and an inhibitor who underwent ITI, and developed NS 19 months into this therapy. A percutaneous renal biopsy was safely performed with factor IX (FIX) concentrate administration both preceding and following the procedure. The patient's inhibitor level had increased to 1.4-1.6 Bethesda Units just prior to the onset of proteinuria. Histology confirmed segmental membranous nephropathy (MGN). The patient was continued on FIX concentrate as ITI and also received 4 weekly doses of rituximab and ongoing immunosuppression with mycophenolate mofetil. This resulted in the complete resolution of his inhibitor and his NS. He continues with a modified ITI regimen and remains inhibitor-free without proteinuria >12 months post-biopsy. CONCLUSIONS: Hemophilia B patients undergoing ITI should be regularly screened for NS. At first detection of proteinuria, with proper precautions, a percutaneous kidney biopsy can be performed safely in patients with low levels of inhibitor. Our patient had segmental MGN with complete remission of NS.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies/blood , Coagulants/immunology , Factor IX/immunology , Glomerulonephritis, Membranous/therapy , Hemophilia B/drug therapy , Immunosuppressive Agents/administration & dosage , Mycophenolic Acid/analogs & derivatives , Nephrotic Syndrome/therapy , Biopsy , Child, Preschool , Coagulants/adverse effects , Coagulants/antagonists & inhibitors , Factor IX/adverse effects , Factor IX/antagonists & inhibitors , Glomerulonephritis, Membranous/diagnosis , Glomerulonephritis, Membranous/immunology , Hemophilia B/blood , Hemophilia B/immunology , Humans , Immune Tolerance , Male , Mycophenolic Acid/administration & dosage , Nephrotic Syndrome/diagnosis , Nephrotic Syndrome/immunology , Proteinuria/immunology , Proteinuria/therapy , Rituximab , Time Factors , Treatment Outcome
17.
Haemophilia ; 18 Suppl 4: 43-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22726082

ABSTRACT

Inhibitor development remains a challenge to appropriate haemophilia treatment. This challenge is being addressed, in part, by an expanding knowledge of the mechanisms that drive inhibitor development including how elements of the innate immune response play a role in inhibitor development. There are promising therapies that may suppress an active immune response. Models to assess the immune responses are becoming ever more sophisticated. Newer models can be used at the preclinical level to evaluate the role of MHC-class II presentation of antigens in both in vitro cell culture studies and in vivo in transgenic mice that express either the protein to be studied or that express human MHC-class II proteins. Parallel to work designed to reduce or reverse inhibitors is development of improved therapies including bypassing agents to treat patients with inhibitors. With these new treatment modalities comes the problem of assessing efficacy at the preclinical level. Models to evaluate bleeding are being developed that may give a more subtle assessment of bypassing agents. These models represent in part an attempt to incorporate the role of ongoing bleeding into the evaluation. Overall, these newer models have great potential in preclinical studies to evaluate the risk of inhibitor development of new therapeutics and to assess the functionality of these new therapeutics.


Subject(s)
Coagulants/therapeutic use , Disease Models, Animal , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Hemophilia A/immunology , Immune Tolerance , Animals , Antigens, CD/analysis , Antigens, CD/immunology , Blood Coagulation Factor Inhibitors/immunology , Coagulants/immunology , Dogs , Factor VIII/immunology , Humans , Mice
18.
Haemophilia ; 18(3): e323-30, 2012 May.
Article in English | MEDLINE | ID: mdl-22044430

ABSTRACT

Haemophilia A is a life long bleeding disorder caused by an inherited deficiency of factor VIII (FVIII). About 30% of haemophilia A patients develop neutralizing antibodies as a consequence of treatment with FVIII concentrates. Immune tolerance protocols for the eradication of inhibitors require daily delivery of intravenous FVIII. We evaluated the immune responses to serial intravenous administration of FVIII in preimmunized haemophilia A mice. We introduced an implantable venous-access device (iVAD) system into haemophilia A mice to facilitate sequential infusion of FVIII. After preimmunization with FVIII, the haemophilia A mice were subjected to serial intravenous administration of FVIII through the iVAD system. In all mice with serial infusion of FVIII, high titers of anti-FVIII inhibitory antibodies developed at 10 exposure days (EDs). However, the anti-FVIII IgG titers were decreased after 150 EDs of sequential low-dose infusion of FVIII [0.05 U g(-1) body weight (BW) five times per week]. Proliferative response to ex vivo FVIII stimulation was significantly suppressed in splenic CD4(+) T cells from mice with serial low-dose FVIII infusion compared with those from mice with high-dose FVIII infusion (0.5 U g(-1) BW five times per week) or preimmunized mice. Moreover, splenic CD4(+) T cells from mice with serial low-dose infusion of FVIII failed to produce interleukin-2 and interferon-γ. These data suggest that serial infusion of FVIII could induce T-cell anergy in haemophilia A mice with inhibitor antibodies.


Subject(s)
Blood Coagulation Factor Inhibitors/immunology , Coagulants/immunology , Factor VIII/immunology , Hemophilia A/immunology , Immune Tolerance/drug effects , Animals , Blood Coagulation Factor Inhibitors/blood , Catheterization, Central Venous , Catheters, Indwelling , Cell Proliferation/drug effects , Coagulants/administration & dosage , Cytokines/metabolism , Disease Models, Animal , Factor VIII/administration & dosage , Hemophilia A/drug therapy , Hemophilia A/metabolism , Immunoglobulin G/blood , Infusions, Intravenous , Isoantibodies/blood , Mice
19.
Haemophilia ; 18 Suppl 4: 48-53, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22726083

ABSTRACT

The management of patients with inhibitors is the greatest challenge facing haemophilia health professionals. Immune tolerance induction (ITI) can be successful in eliminating the inhibitor in the majority of patients, provided it is started soon after the inhibitor develops and the titre of the inhibitor is <10 BU at commencement of ITI. Acute bleeding is treated using one of two bypassing agents, which exhibit similar efficacy and safety. Surgery in inhibitor patients is challenging and should only be carried out in experienced centres.


Subject(s)
Blood Coagulation Factor Inhibitors/blood , Coagulants/therapeutic use , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Immune Tolerance/drug effects , Acute Disease , Coagulants/immunology , Factor VIII/immunology , Hemophilia A/immunology , Hemorrhage/prevention & control , Humans , Immune Tolerance/immunology , Surgical Procedures, Operative/methods
20.
Haemophilia ; 18 Suppl 5: 6-10, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22757678

ABSTRACT

Animal models have played a critical role in developing our understanding of haemophilia and its treatment. For example, studies in mice and dogs have provided insights into the pharmacokinetics and pharmacodynamics of recombinant activated factor VII (rFVIIa). Such studies have shown that antithrombin has a significant impact on clearance of rFVIIa, which explains discrepancies between the antigen and activity half-lives of rFVIIa. Animal studies have also shown that the major clearance organs for rFVIIa are the liver and the kidneys, whereas distribution studies suggest that FVII and rFVIIa leave the circulation and enter the tissues, before returning to the circulation through the lymph. One agent that has benefited greatly from the use of animal models in its development is vatreptacog alfa, a new analogue of rFVIIa. Promising in vitro results, including increased generation of FXa, shortened clotting times and increased clot stability, were subsequently confirmed in animal models. In a severe tail-bleed model in FVIII knock-out mice, reduction in maximal blood loss was substantially greater with vatreptacog alfa than with rFVIIa, FVIII or plasma-derived activated prothrombin complex concentrate. In a mouse model of joint bleeding, rFVIIa and vatreptacog alfa significantly reduced bleeding compared with vehicle-treated haemophilic controls. More recently, a model of endothelial injury based on mouse cremaster muscle has been developed. Overall, animal models are a valuable tool in elucidating the haemostatic process and the effects of therapeutic agents, although direct extrapolation to the clinical setting should be done with caution.


Subject(s)
Coagulants/pharmacokinetics , Factor VIIa/pharmacokinetics , Hemophilia A/drug therapy , Animals , Antigens/blood , Blood Coagulation/drug effects , Coagulants/immunology , Coagulants/therapeutic use , Disease Models, Animal , Dogs , Factor VIIa/immunology , Factor VIIa/therapeutic use , Half-Life , Hemophilia A/immunology , Kidney/metabolism , Liver/metabolism , Mice , Rats , Recombinant Proteins/immunology , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Tail/blood supply
SELECTION OF CITATIONS
SEARCH DETAIL