Induction of Tolerance to FVIII in Hemophilic Mice by Delivery of Apoptotic Syngeneic Fibroblasts Expressing a FVIII Transgene.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 768-768
Author(s):  
Rui-Jun Su ◽  
Laura Stewart ◽  
Angela Epp ◽  
Steven W. Pipe ◽  
Neil C. Josephson

Abstract In response to treatment with factor concentrates approximately 30% of patients with severe hemophilia A develop high titer inhibitory antibodies to Factor VIII. Inhibitor formation represents a major obstacle in treating patients of hemophilia A. We are investigating ways to prevent and treat inhibitors, in the mouse model of hemophilia A, through tolerance induction by infusion of FVIII vector modified apoptotic syngeneic fibroblasts. We generated a fibroblast cell line from a tail snip of a 129Sv−FVIIIKO mouse. We then transduced these cells with a bi−cistronic foamy virus vector expressing both the full length FVIII coding sequence and the zeocin resistance gene. Production of FVIII by these modified cells was confirmed by ELISA on cell culture medium. A cell line transduced with a control vector expressing only the zeocin resistance gene was also generated. Both vector modified cell lines were maintained under selective pressure and induced to apoptose by UV irradiation just prior to infusion into 129Sv−FVIIIKO mice. Mice were treated with two infusions of UV−irradiated FVIII expressing or control fibroblasts at three different weekly cell doses: 1 x 107, 2 x 106, or 2 x 105. Ten days later they were inoculated with 4 weekly intravenous doses of 0.2 μg albumin free recombinant human FVIII (ADVATE). An additional set of control mice were not given any apoptotic cells prior to inoculation with ADVATE. Blood was collected to determine anti−FVIII inhibitor titers one week after the final ADVATE dose. The Bethesda titers in mice treated with apoptotic FVIII expressing fibroblasts, at all 3 cell doses, were 3–5 fold lower than mice that received no cells prior to intravenous FVIII challenge (p < 0.02 for all 3 cohorts, Welch’s t−test). A pair wise comparison with mice that received equivalent numbers of control apoptotic cells demonstrated significantly lower Bethesda titers in the mice given two weekly doses of 2 x106 FVIII expressing cells (436 ± 140BU versus 1230 ± 433BU, p = 0.04). In mice that received 1 x 107 and 2 x 105 apoptotic FVIII expressing cells there was a trend for the development of lower Bethesda titers when compared to mice that received an equivalent numbers of apoptotic cells modified by the control vector. Four months after treatment with ADVATE mice from the no cell group and both the 1 x 107 control and FVIII vector expressing groups were re−challenged with 4 additional weekly doses of ADVATE. T cell proliferation assays showed the highest stimulation index in mice that received no cells and the lowest index in mice that were treated with apoptotic FVIII expressing fibroblasts. Bethesda titer results from these re−challenged mice are currently pending. Our data show that anti−Factor VIII inhibitory antibody titers can be reduced by the prior infusion of apoptotic syngeneic cells stably transduced with a FVIII expressing foamy virus vector. Furthermore, the higher titers seen in mice treated with cells containing a control vector demonstrate that the effect is dependent on delivery of FVIII within the apoptotic cells. Moreover, the T cell stimulation data seen in the mice re−challenged 4 months after initial inoculation with FVIII indicate that the decrease in immune priming is due to the induction of durable tolerance. Future work will focus on elucidating the mechanisms of tolerance induction in this model and on combining apoptotic cell delivery with other immune modulators to optimize results.

Author(s):  
Н.И. Зозуля

Серьезным осложнением, связанным с лечением гемофилии А, является развитие ингибиторов. В последние годы был проведён ряд исследований, посвящённых данной проблеме: RODIN, INSIGHT, FranceCoag, SIPPET и NuProtect. В данном обзоре суммируются основные результаты этих исследований. Согласно результатам рандомизированного исследования SIPPET, препараты плазматического фактора свертывания крови VIII (FVIII) обладают меньшей иммуногенностью, чем препараты рекомбинантного FVIII, синтезированного из клеточной линии китайских хомячков, что следует учитывать при выборе стратегии лечения. Согласно результатам исследования NuProtect, опубликованным в 2019 г., концентрат рекомбинантного FVIII, полученный из клеточной линии человека, демонстрирует профиль иммуногенности, сходный с таковым у препаратов плазматического FVIII. У ранее нелеченых пациентов с ненулевыми мутациями при применении симоктоког альфа не наблюдалось образования ингибиторов, также как и в случае применения препаратов плазматического FVIII в исследовании SIPPET. Inhibitor development is a serious complication associated with hemophilia A therapy. A number of studies have been carried out of this issue — RODIN, INSIGHT, FranceCoag, SIPPET, and NuProtect. This review summarizes the main results of these studies. According to the results of the SIPPET randomized trial, plasma-derived coagulation factor VIII (FVIII) products are less immunogenic than recombinant FVIII products synthesized from a Chinese hamster cell line; this fact should be taken into account in choosing a treatment strategy. According to the results of NuProtect study published in 2019, the concentrate of human cell line-derived recombinant FVIII demonstrates immunogenicity profi le similar to the one in plasma-derived FVIII products. Previously untreated patients with non-zero mutations receiving simoctocog alfa did not show development of inhibitors as well as in case of administration of plasma-derived FVIII products in SIPPET study.


Blood ◽  
1990 ◽  
Vol 75 (2) ◽  
pp. 378-383 ◽  
Author(s):  
IM Nilsson ◽  
E Berntorp ◽  
O Zettervall ◽  
B Dahlback

Abstract We recently described tolerance induction with factor VIII/IX, cyclophosphamide, and high-dose intravenous IgG in hemophilia A or B patients with coagulation inhibitory antibodies. Circulating noninhibitory antibodies complexed with factor IX have been demonstrated in tolerant hemophilia B patients. Similar findings are now described in six tolerant hemophilia A patients. Complexes between factor VIII and the ‘tolerant’ antibody were demonstrated by subjecting plasma to gel filtration chromatography, void fractions containing factor VIII/vWF complexes being collected and adsorbed to protein A. Using 125I-labeled F(ab')2 fragments against IgG subclass and factor VIII antigen, complexes between an IgG4 antibody and factor VIII were found to adsorb to protein A. After infusion of factor VIII to tolerant patients, all factor VIII circulated in complex with IgG4 antibody. In three of the patients, the ‘tolerant’ antibodies inhibited an ELISA specific for factor VIII light chain but, unlike the pretolerant antibodies, did not bind radiolabeled factor VIII heavy chain. Although after induction of tolerance the patients still have circulating IgG4 antibodies against factor VIII, the antibodies differ in specificity, lack coagulation inhibitory activity, and do not enhance the rate of elimination of factor VIII.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 216-216
Author(s):  
Rui-Jun Su ◽  
Laura Stewart ◽  
Steven W. Pipe ◽  
Neil C. Josephson

Abstract Approximately 30% of patients with severe hemophilia A develop neutralizing antibodies (inhibitors) to factor VIII (FVIII). Frequently, the inhibitors that develop are persistent and of sufficiently high titer that infusion of FVIII concentrates are ineffective for the control of bleeding episodes. At present the only method for elimination of high titer inhibitors is Immune Tolerance Induction (ITI) by exposing patients to repeated FVIII doses. However, it is extremely expensive and takes many months to complete. There is a need to develop quicker, cheaper, and more reliable protocols for inducing tolerance in patients with high titer FVIII inhibitors, and of preventing their occurrence in patients at high risk. In a non-inflammatory environment, autologous apoptotic cells are processed by immature, non-activated dendritic cells (DCs) capable of initiating peripheral immune tolerance through the stimulation of different classes of regulatory T cells. Furthermore, it has been demonstrated that antigen specific tolerance can be induced by delivery of a foreign protein within dying syngeneic cells (Liu et al, J Exp Med196: 1091, 2002). We hypothesize that immune tolerance to FVIII can be induced by the infusion of apoptotic autologous cells engineered to carry a FVIII expression vector. We generated a fibroblast cell line from a tail snip of a 129Sv-FVIIIKO mouse and transduced the cells with a foamy virus vector expressing a B-domain deleted human FVIII construct. Expression of human FVIII was detected by ELISA in the supernatant of cultured transduced fibroblasts. These transduced cells were induced to apoptosis by serum starvation for 24–30 hr prior to infusion into 129Sv-FVIIIKO mice. Treated mice received two weekly doses of 1x107 serum starved transduced fibroblasts. Control mice were infused with culture media alone. Ten days after the final infusion, experimental and control mice were challenged with 4 weekly intravenous doses of 0.2 μg albumin free recombinant human FVIII (ADVATE). Ten days after the final dose of ADVATE blood samples were collected and evaluated for inhibitor titer by Bethesda assay and for total anti-FVIII antibody titer by ELISA. Three weeks later, T cell proliferation assays were performed on samples from mice in each group. The mice that received apoptotic cells had lower inhibitor titers than controls, 156.7 ± 82.7 BU/ml (n = 6) versus 331.3 ± 183.9 BU/ml (n = 11) (p = 0.017, Welch’s t-test). However, total antibody titer levels determined by ELISA were not significantly different between the two groups. T-cell proliferation assays also showed a reduced T cell response in the apoptotic cell treated mice with a stimulation index that was half that seen in the controls. Our data show that inhibitor titers and T cell responses in hemophilia A mice challenged by albumin free recombinant FVIIII can be reduced by the prior infusion of apoptotic syngeneic cells transduced with a human FVIII expressing vector. Future work will focus on optimizing apoptotic cell delivery protocols to maximally suppress the immune response to FVIII and on elucidating the mechanisms responsible for our findings. Figure Figure


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1279-1279
Author(s):  
Peiqing Ye ◽  
David J. Rawlings ◽  
Arthur R. Thompson ◽  
Hans D. Ochs ◽  
Carol H. Miao

Abstract Naked DNA transfer of liver-specific, high-expressing plasmid pBS-HCRHPI-FVIIIA in Rag2(−/ −) SCID mice produced persistent high-level gene expression of human factor VIII (hFVIII) (Miao, Hum. Gene Ther. 2003). However, in immunocompetent hemophilia A mice, a robust humoral immune response against FVIII that followed gene transfer led to complete inhibition of circulating FVIII activity (Ye, Mol. Ther. 2004). Transient immunomodulation strategies were explored to prevent the formation of inhibitory antibody formation. Eight groups of mice (n=8) were treated by naked DNA transfer of plasmid pBS-HCRHPI-FVIIIA. Each group were subjected to treatment with single or combined immunosuppressive regimen: CyclosporineA (CSA) daily for 14 days; Rapamycin daily for 14 days; Mycophenylate mofetil (MMF) daily for 14 days; combination of CSA and MMF; combination of Rapamycin and MMF; a monoclonal antibody (MR1) against murine CD40 ligand on days -1, 1, 2, 7, & 14; recombinant murine Ctla4Ig on days 1 & 2; and combination of MR1 and Ctla4Ig. Combination regimens were given using the same combined schedule and dosages. All animals treated with immunosuppression had delayed or no immune responses against hFVIII except the group treated with CSA only. The most effective treatment was observed in animals treated with the combination of Ctla4Ig and MR1. Seven of 8 animals failed to develop detectable inhibitors. One animal developed transient low-titer antibodies. This group of animals produced persistent, therapeutic levels of hFVIII gene expression for over 6 months. Tolerized animals were subsequently challenged by the T dependent antigen, bacteriophage Φx174, and exhibited a normal primary and secondary response including amplification and isotype switch. These results strongly suggest that transient immunomodulation strategies to disrupt B- and T- cell interactions at the time of plasmid injection is effective to promote long-term immune tolerance that is specific for FVIII without altering subsequent immune responses to other T cell dependent antigens.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4074-4074 ◽  
Author(s):  
Chirag J. Amin ◽  
Alice D. Ma

Abstract Inhibitor development in congenital hemophiliacs can be clinically catastrophic. Immune tolerance induction therapy has previously been the standard of care in eradicating inhibitors; however due to a multitude of factors, this may not be applicable in certain patients. The role of Rituximab is receiving more attention in this subset of patients. In this abstract, we report that treatment with Rituximab led to successful eradication of high-titer inhibitors in 3 patients with mild to moderate hemophilia A who developed inhibitors after receiving intensive treatment with recombinant Factor VIII (FVIII). Patient Characteristics: Three patients, aged 50–70, with baseline FVIII levels of 2–9%, developed inhibitors after recombinant Factor VIII infusion. Patient A was treated with continuous infusion FVIII for a post-surgical hemarthrosis for approximately 7 days. Patient B received bolus dose FVIII for a GI bleed for at least 10 days, and Patient C received bolus dose FVIII for knee replacement for 10 days. Factor VIII inhibitors were detected in these patients after one month. None of these patients had been treated with immune tolerance previously or had known inhibitors. Each patient received Rituximab 375mg/m2 every week for 4 weeks total. During and after treatment, FVIII levels and Bethesda inhibitor titers (BU) were monitored. Results: All three patients had eradication of their inhibitors (Figure 1) and return of their FVIII levels to baseline by six months post-treatment. Notably, patient C’s inhibitor peak was 117 BUs, 7 months prior to Rituximab treatment. Patient C’s initial response to Rituximab has been previously reported at ASH in abstract form. We now report that 4 years later, this patient has had a recurrence of his inhibitor after monoclonal FVIII for a contralateral knee replacement but with a peak titer of only 2 (Table 1). Inhibitor Trends after Rituximab Treatment Inhibitor Trends after Rituximab Treatment Bethesda Inhibitor Titer (BU) per Month (*) after Receiving Rituximab 0* 1 3 6 36 48 51 NA=Not applicable as data has not matured yet Patient A (BU) 5 0.7 0 0 NA NA NA Patient B (BU) 17 7 2 0 NA NA NA Patient C (BU) 40 4 0 0 0 2 0.5 Conclusion: Inhibitors in patients with mild-moderate hemophilia differ from those with severe FVIII deficiency, behaving more like the autoantibodies seen in patients with spontaneous FVIII inhibitors. In support of this idea, we successfully treated high titer inhibitors which developed in 3 patients with baseline FVIII levels of 2–9%. All three patients had prompt resolution of their inhibitor titers during the course of therapy, with return of their baseline FVIII levels. Historically, patients with mild-moderate hemophilia treated at the Harold R. Roberts Comprehensive Hemophilia Center at the University of North Carolina were treated either with immune tolerance induction or by bypass agents alone, with inhibitor eradication taking months to years (data not shown). While performance of larger prospective trials would be ideal, the small number of patients with this condition limits the ability to perform these trials. Our findings, in combination with other case series from other institutions, reveal a promising alternative for prompt and reliable treatment in mild-moderate hemophiliacs with inhibitors.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3526-3526
Author(s):  
David W. Scott ◽  
Elizabeth Kadavil ◽  
Ai-Hong Zhang ◽  
Ruth A. Ettinger ◽  
Kathleen Pratt

Abstract A major obstacle in the treatment of Hemophilia A is that patients can develop an inhibitory immune response to therapeutic doses of coagulation factor VIII (fVIII). Over the last decade, we have developed a B-cell delivered gene therapy approach to prevent the development of inhibitory antibodies (“inhibitors”) in fVIII knockout mice (see Lei and Scott, Blood105: 4865, 2005). In our murine platform, activated primary spleen B cells or bone marrow cells are transduced with a retroviral vector encoding the fVIII A2 and/or C2 domain fused to an IgG heavy chain, and these cells are injected systemically into immunocompetent fVIII knockout animals. The recipients are rendered specifically tolerant to the encoded C2 and A2 domains, as evidenced by a >90% reduction of inhibitor titers, even in primed animals. To help evaluate the potential of this approach for translation, we are developing in vitro models for tolerance induction using human T-cell clones isolated from subjects with mild hemophilia A. The clones are isolated by single-cell sorting of CD4+ cells that are labeled by fluorescent HLA-DR tetramers complexed with peptides containing fVIII epitopes, followed by expansion with HLA-DR mismatched peripheral blood mononuclear cells (PBMC), phytohaemagglutinin, and interleukin-2. Our initial model utilizes a T-cell clone from an individual with mild hemophilia A due to fVIII missense genotype A2201P, which recognizes an HLA-DRA-DRB1*0101-restricted epitope within a synthetic peptide corresponding to fVIII residues 2194–2213. All of the antigen-specific T-cell clones isolated from this subject secreted interferon-gamma (IFN-γ) when stimulated by fVIII2194–2213 presented by irradiated HLA-DR-matched PBMCs or with plate-bound anti-CD3. Because of their robust response to a clinically relevant epitope in fVIII, one of these clones that expanded well in culture was chosen for initial testing of a modified gene therapy platform similar to that developed using the murine hemophilia A model. HLA-matched peripheral blood B cells were activated with antibodies to IgM or with CD40L-expressing fibroblasts and then transduced with a modified retroviral vector containing the human C2 domain sequence in-frame with the IgG sequence. These B cells were cultured with the hemophilic T-cell clone. After pre-treatment (“tolerance-induction step”), the cells were washed and then stimulated by plate-bound anti-CD3. The subsequent IFN-γ response (measured by ELIspots and ELISA) was dramatically reduced compared to the response of same T-cell clone cultured with mock-transduced B cells. The post-treatment reduction in IFN-γ secretion was equivalent to that induced after soluble anti-CD3 pre-treatment, a known method to induce T-cell anergy in vitro. Interestingly, IL-10 was produced during the tolerance induction (pre-treatment) phase, most likely from the activated B cells. Preliminary, parallel experiments with B cells transduced with a “gutless” adenovirus vector expressing C2-Ig did not result in a similar down-regulation of the T-cell response, suggesting that this non-integrating method of expressing antigens for tolerance is not effective, at least in this system. These results are the first to demonstrate in vitro modulation of cytokine responses using DR-restricted, fVIII-specific T cells from a hemophilia A subject. Further investigations using T-cell clones from hemophilic subjects with and without anti-fVIII antibodies will allow us to explore mechanisms of tolerance and may also suggest novel approaches to reduce inhibitor titers.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5050-5050
Author(s):  
Mark J. Belletrutti ◽  
Roxanne Seiferman-Nelson ◽  
Bonny Granfield

Abstract Introduction: Development of circulating anti-factor VIII antibodies (inhbitors) is the most serious and challenging complication in the treatment of hemophilia A. Up to 38% of hemophilia patients develop inhibitors with recombinant FVIII (rFVIII) products (Gouw et al. N Engl J Med. 2013; 368:231-239). The presence of inhibitors leads to an increased risk of bleeding, poor physical functioning and quality of life (Benson et al., Eur. J. Haematol. 2012; 88:371-379). Immune tolerance induction (ITI) is the most common method for eliminating inhibitors, historically performed with high dose, and prolonged treatment with plasma-derived (pd), or recombinant FVIII (rFVIII) concentrates. Although ITI for the eradication of inhibitors has become standard of care for hemophilia patients the therapeutic superiority of a particular product type (rFVIII vs. pd-FVIII) has not yet been conclusively demonstrated. In accordance with its role in stabilizing FVIII, the presence of von Willebrand factor (VWF) in pd-FVIII concentrates has been shown to improve the outcome of ITI. Wilate® (Octapharma) is a high-purity human plasma derived complex containing two proteins (VWF and FVIII) in a 1:1 ratio. The aim of this study was to determine the effectiveness of Wilate for primary ITI therapy for six patients with severe hemophilia A. Patients and Methods: The case history for six pediatric hemophilia A patients prior to and during primary Wilate ITI was reviewed. For 5/6 patients, inhibitors developed during rFVIII factor replacement therapy. For the sixth patient, inhibitors were detected at the time of hemophilia diagnosis. ITI began once patients achieved an inhibitor titer of less than 10 BU/mL. The ITI dosing regimen ranged from 50-60 IU/Kg of Wilate three times per week to 200 IU/Kg once daily. Inhibitor titers were measured regularly, prior to and during ITI using the Nijmegen-Bethesda assay. The number of port-a-cath infections and bleeding episodes were also monitored. ITI success was defined as: an undetectable inhibitor level (<0.6 BU/mL), FVIII plasma recovery ≥ 66% of predicted, and FVIII half-life ≥6 hours. Results: Wilate ITI was well tolerated in all patients, with no product-related adverse events. All patients had a port-a-cath device inserted for Wilate injections. Two port-a-cath infections occurred during ITI. Five of six patients had poor prognostic factors for ITI outcome. These poor prognostic factors included a high-risk FVIII gene mutation, historical peak inhibitor titer greater than 50 BU/mL, age of ITI onset greater than 6 years, and ITI onset more than 12 months from inhibitor development. The frequency of these poor prognostic factors varied amongst the patients: 1 patient had 4, 1 patient had 2, and 3 patients presented with 1 poor prognostic factor. Despite the presence of these high-risk factors, Wilate was successful at reducing the inhibitor titers to undetectable levels in all patients. Furthermore, inhibitor titers have remained low or undetectable without significant spikes for the duration of treatment. Patient plasma recovery and FVIII half-life results have also indicated that patients are progressing towards successful ITI. Importantly, for 6/6 patients (including 3 patients who had previously been treated with Anti-Inhibitor Coagulant Complex (FEIBA) prophylaxis therapy) - Wilate therapy was successful at reducing the number of bleeding episodes allowing for the cessation of FEIBA prophylaxis. Since commencing Wilate ITI, 6/6 patients have not reported any major bleeding episodes. The improved clinical outcome was perceived by the patients as an improved well-being, and quality of life. Conclusion: Wilate ITI was found to be well tolerated, safe, and successful at reducing inhibitor levels to below the detectable range for six severe hemophilia A patients. Patients experienced no treatment related adverse events, had a low rate of port-a-cath infections, and did not present with any major bleeding episodes while on Wilate ITI. In light of the 3-5 fold increase in overall treatment costs of immune tolerance induction, careful consideration should be given to choice of product (rFVIII versus pd-FVIII) – especially for patients at high-risk of failure. (Dimichele et al. Haemophilia 2004: 10 Suppl 4;140-145). The present data suggest that Wilate, a pd-FVIII product, is effective in managing patients with inhibitors. Disclosures Belletrutti: Baxter Canada: Honoraria, Membership on an entity's Board of Directors or advisory committees; CSL Behring Canada: Honoraria.


Blood ◽  
2012 ◽  
Vol 119 (18) ◽  
pp. 4108-4114 ◽  
Author(s):  
Pier Mannuccio Mannucci ◽  
Maria Elisa Mancuso ◽  
Elena Santagostino

AbstractIn high-income countries, the large availability of coagulation factors for replacement therapy of patients with hemophilia A has raised the life expectancy of these lifelong bleeders to that of males from the general population. The practicing clinician is offered a multitude of choices among several commercial brands of factor VIII extracted from human plasma or engineered from mammalian cell cultures by means of recombinant DNA technology. This article has the goal to offer our opinions on how to choose among the different products, that we consider interchangeable relevant to their clinical efficacy in the control of bleeding and safety from pathogen transmission. Hence, the main determinants of our choices are price and the risk of occurrence of factor VIII inhibitory alloantibodies. With this as background, we present the rationale underlying the choices for different categories of patients with severe hemophilia A: previously untreated patients, multiply treated patients, and patients undergoing immune tolerance induction with large doses of factor VIII to eradicate inhibitors. Mention is also made to the possible strategies that should be implemented to make available coagulation factors for replacement therapy in developing countries.


Blood ◽  
2013 ◽  
Vol 121 (22) ◽  
pp. 4449-4456 ◽  
Author(s):  
David W. Scott ◽  
Kathleen P. Pratt ◽  
Carol H. Miao

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.


Sign in / Sign up

Export Citation Format

Share Document