Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Exp Immunol ; 204(2): 258-266, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33512707

RESUMO

The mechanisms of action of intravenous immunoglobulins (IVIg) in autoimmune diseases are not fully understood. The fixed duration of efficacy and noncumulative effects of IVIg in immune thrombocytopenia (ITP) and acquired von Willebrand disease (AVWD) suggest other mechanisms besides immunological ones. Additionally to the peripheral destruction of platelets in ITP, their medullary hypoproduction emerged as a new paradigm with rescue of thrombopoietin receptor agonists (TPO-RA). In an ITP mouse model, interleukin (IL)-11 blood levels increase following IVIg. IL-11 stimulates the production of platelets and other haemostasis factors; recombinant IL-11 (rIL-11) is thus used as a growth factor in post-chemotherapy thrombocytopenia. We therefore hypothesized that IVIg induces IL-11 over-production, which increases platelets, VWF and factor VIII (FVIII) levels in humans and mice. First, in an ITP mouse model, we show that IVIg or rIL-11 induces a rapid increase (72 h) in platelets, FVIII and VWF levels, whereas anti-IL-11 antibody greatly decreased this effect. Secondly, we quantify for the first time in patients with ITP, AVWD, inflammatory myopathies or Guillain-Barré syndrome the dramatic IL-11 increase following IVIg, regardless of the disease. As observed in mice, platelets, VWF and FVIII levels increased following IVIg. The late evolution (4 weeks) of post-IVIg IL-11 levels overlapped with those of VWF and platelets. These data may explain thrombotic events following IVIg and open perspectives to monitor post-IVIg IL-11/thrombopoietin ratios, and to assess rIL-11 use with or without TPO-RA as megakaryopoiesis co-stimulating factors to overcome the relative hypoproduction of platelets or VWF in corresponding autoimmune diseases, besides immunosuppressant.


Assuntos
Plaquetas/imunologia , Fator VIII/imunologia , Imunoglobulinas Intravenosas/imunologia , Interleucina-11/imunologia , Fator de von Willebrand/imunologia , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Estudos Retrospectivos
2.
Haemophilia ; 22 Suppl 5: 31-5, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27405673

RESUMO

INTRODUCTION: Haemophilia A is an X-linked bleeding disorder characterized by a deficiency of coagulation protein factor VIII (FVIII). A challenging complication of therapeutic FVIII infusions is the formation of neutralizing alloantibodies against the FVIII protein defined as inhibitors. The development of FVIII inhibitors drastically alters the quality of life of the patients and is associated with tremendous increases in morbidity as well as treatment costs. AIM: Current clinical immune tolerance induction protocols to reverse inhibitors are lengthy, costly and not effective in all patients. Prophylactic protocols to prevent inhibitor formation have not yet been developed in the clinical setting. However, there has been ample progress towards this goal in recent years in preclinical studies using animal models of haemophilia. METHODS: Here, we review the mechanisms that lead to inhibitor formation against FVIII and two promising new strategies for antigen-specific tolerance induction. RESULTS: CD4+ T cells play an important role in the FVIII-specific B cell response. Immune tolerance can be induced based on transplacental delivery of FVIII domains fused to Fc or on oral delivery of leaf cells from chloroplast transgenic crop plants. CONCLUSIONS: Recent literature suggests that prophylactic tolerance induction protocols for FVIII may be feasible in haemophilia A patients.


Assuntos
Hemofilia A/imunologia , Tolerância Imunológica , Animais , Anticorpos Neutralizantes/sangue , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Modelos Animais de Doenças , Fator VIII/imunologia , Fator VIII/uso terapêutico , Feminino , Hemofilia A/tratamento farmacológico , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Troca Materno-Fetal , Plantas Geneticamente Modificadas/imunologia , Gravidez
3.
Haemophilia ; 22 Suppl 1: 1-24, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26728503

RESUMO

Over the past 20 years, there have been many advances in haemophilia treatment that have allowed patients to take greater control of their disease. However, the development of factor VIII (FVIII) inhibitors is the greatest complication of the disease and a challenge in the treatment of haemophilia making management of bleeding episodes difficult and surgical procedures very challenging. A meeting to discuss the unmet needs of haemophilia patients with inhibitors was held in Paris on 20 November 2014. Topics discussed were genetic and non-genetic risk factors for the development of inhibitors, immunological aspects of inhibitor development, FVIII products and inhibitor development, generation and functional properties of engineered antigen-specific T regulatory cells, suppression of immune responses to FVIII, prophylaxis in haemophilia patients with inhibitors, epitope mapping of FVIII inhibitors, current controversies in immune tolerance induction therapy, surgery in haemophilia patients with inhibitors and future perspectives for the treatment of haemophilia patients with inhibitors. A summary of the key points discussed is presented in this paper.


Assuntos
Anticorpos/imunologia , Fator VIII/imunologia , Fator VIII/uso terapêutico , Hemofilia A/imunologia , Hemofilia A/terapia , Mapeamento de Epitopos , Fator VIII/genética , Hemofilia A/genética , Hemofilia A/cirurgia , Humanos , Tolerância Imunológica , Imunidade Celular , Paris
4.
Clin Exp Immunol ; 181(3): 385-400, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25959571

RESUMO

Biopharmaceuticals (BPs) represent a rapidly growing class of approved and investigational drug therapies that is contributing significantly to advancing treatment in multiple disease areas, including inflammatory and autoimmune diseases, genetic deficiencies and cancer. Unfortunately, unwanted immunogenic responses to BPs, in particular those affecting clinical safety or efficacy, remain among the most common negative effects associated with this important class of drugs. To manage and reduce risk of unwanted immunogenicity, diverse communities of clinicians, pharmaceutical industry and academic scientists are involved in: interpretation and management of clinical and biological outcomes of BP immunogenicity, improvement of methods for describing, predicting and mitigating immunogenicity risk and elucidation of underlying causes. Collaboration and alignment of efforts across these communities is made difficult due to lack of agreement on concepts, practices and standardized terms and definitions related to immunogenicity. The Innovative Medicines Initiative (IMI; www.imi-europe.org), ABIRISK consortium [Anti-Biopharmaceutical (BP) Immunization Prediction and Clinical Relevance to Reduce the Risk; www.abirisk.eu] was formed by leading clinicians, academic scientists and EFPIA (European Federation of Pharmaceutical Industries and Associations) members to elucidate underlying causes, improve methods for immunogenicity prediction and mitigation and establish common definitions around terms and concepts related to immunogenicity. These efforts are expected to facilitate broader collaborations and lead to new guidelines for managing immunogenicity. To support alignment, an overview of concepts behind the set of key terms and definitions adopted to date by ABIRISK is provided herein along with a link to access and download the ABIRISK terms and definitions and provide comments (http://www.abirisk.eu/index_t_and_d.asp).


Assuntos
Hipersensibilidade a Drogas/prevenção & controle , Drogas em Investigação/normas , Guias como Assunto/normas , Terminologia como Assunto , Alergia e Imunologia/normas , Hipersensibilidade a Drogas/imunologia , Indústria Farmacêutica/organização & administração , Indústria Farmacêutica/normas , Drogas em Investigação/efeitos adversos , Drogas em Investigação/uso terapêutico , Humanos , Inovação Organizacional , Política Organizacional , Padrões de Referência
8.
J Thromb Haemost ; 12(12): 2065-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25267332

RESUMO

BACKGROUND: Immune responses to therapeutic factor VIII remain a major problem, affecting 30% of patients with severe hemophilia A. The primary factors that drive immune responses in these patients remain elusive. There have been conflicting reports on a role of coagulation (or thrombin) in anti-FVIII immune responses. OBJECTIVE: To assess the importance of coagulation-associated processes for the onset of the anti-FVIII immune response. METHODS: Using FVIII-deficient mice, we compared the immunogenicity of recombinant FVIII or the inactive FVIII(V) (634M) mutant. In parallel, the involvement of tissue factor (TF) activity in the anti-FVIII immune response was investigated upon injection of a neutralizing anti-TF antibody or by the use of chimeric mice that lack TF expression in myeloid cells. The development of the anti-FVIII immune response was also monitored after treatment with warfarin. RESULTS: The kinetics of the development of antibody responses to FVIII(V) (634M) were indistinguishable from those of wild-type FVIII. Inhibition of TF activity did not modulate immune responses to exogenous FVIII. Additionally, global inhibition of coagulation with warfarin failed to reduce the anti-FVIII immune response. CONCLUSIONS: Thrombin generation or coagulation-associated processes do not modulate the anti-FVIII antibody response in mouse model of severe hemophilia A.


Assuntos
Fator VIII/imunologia , Hemofilia A/sangue , Imunidade Humoral , Animais , Anticorpos Neutralizantes/imunologia , Coagulação Sanguínea , Modelos Animais de Doenças , Hemofilia A/genética , Inflamação , Camundongos , Mutação , Plasmídeos , Estrutura Terciária de Proteína , Proteínas Recombinantes/imunologia , Trombina/química , Tromboplastina/química , Varfarina/farmacologia
9.
Haemophilia ; 20(2): 176-84, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24118514

RESUMO

Forty per cent of haemophilia A (HA) patients have missense mutations in the F8 gene. Yet, all patients with identical mutations are not at the same risk of developing factor VIII (FVIII) inhibitors. In severe HA patients, human leucocyte antigen (HLA) haplotype was identified as a risk factor for onset of FVIII inhibitors. We hypothesized that missense mutations in endogenous FVIII alter the affinity of the mutated peptides for HLA class II, thus skewing FVIII-specific T-cell tolerance and increasing the risk that the corresponding wild-type FVIII-derived peptides induce an anti-FVIII immune response during replacement therapy. Here, we investigated whether affinity for HLA class II of wild-type FVIII-derived peptides that correspond to missense mutations described in the Haemophilia A Mutation, Structure, Test and Resource database is associated with inhibitor development. We predicted the mean affinity for 10 major HLA class II alleles of wild-type FVIII-derived peptides that corresponded to 1456 reported cases of missense mutations. Linear regression analysis confirmed a significant association between the predicted mean peptide affinity and the mutation inhibitory status (P = 0.006). Significance was lost after adjustment on mutation position on FVIII domains. Although analysis of the A1-A2-A3-C1 domains yielded a positive correlation between predicted HLA-binding affinity and inhibitory status (OR = 0.29 [95% CI: 0.14-0.60] for the high affinity tertile, P = 0.002), the C2 domain-restricted analysis indicated an inverse correlation (OR = 3.56 [1.10-11.52], P = 0.03). Our data validate the importance of the affinity of FVIII peptides for HLA alleles to the immunogenicity of therapeutic FVIII in patients with missense mutations.


Assuntos
Fator VIII/genética , Fator VIII/imunologia , Hemofilia A/genética , Hemofilia A/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Isoanticorpos/imunologia , Mutação de Sentido Incorreto , Fragmentos de Peptídeos/imunologia , Sequência de Aminoácidos , Bases de Dados Genéticas , Epitopos/química , Epitopos/imunologia , Fator VIII/química , Hemofilia A/tratamento farmacológico , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Ligação Proteica/imunologia , Fatores de Risco
10.
Haemophilia ; 20(4): 486-92, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24372710

RESUMO

The administration of therapeutic factor VIII (FVIII) to treat or prevent haemorrhages in haemophilia A patients results, in up to 30% of the cases, in the development of inhibitory anti-FVIII antibodies. Much debate has taken place on the relevance of the nature of the FVIII product as a risk factor for inhibitor development. Thus, the plasma-derived vs. recombinant origin, the second vs. third generation of the product, or the presence of the B domain have been controversially evoked. A few years ago, Refacto AF, a third-generation recombinant B domain-deleted FVIII was marketed. The aim of this study was to compare the immunogenicity of Refacto AF to that of two recombinant full-length FVIII products: Helixate and Advate. For the three recombinant FVIII products, we compared the binding to the mannose-sensitive endocytic receptor CD206, the dose-dependent endocytosis by immature monocyte-derived dendritic cells (DCs), the activation by FVIII-loaded DCs of a FVIII-specific HLA-DRB1*0101-restricted mouse T-cell hybridoma and the induction of inhibitory anti-FVIII IgG in FVIII-deficient mice. At elevated FVIII concentrations, Refacto AF was less endocytosed than full-length recombinant products. At lower concentrations, however, Refacto AF was endocytosed by DCs and activated T cells as well as Helixate and Advate. The levels of inhibitory anti-FVIII IgG induced by Refacto AF in FVIII-deficient mice were lower or equal to that induced by Helixate and Advate respectively. The predicted immunogenicity of Refacto AF is identical to or lower than that of the two recombinant full-length FVIII products available on the French market.


Assuntos
Fator VIII/efeitos adversos , Fator VIII/imunologia , Animais , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Endocitose , Fator VIII/metabolismo , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Hemofilia A/imunologia , Humanos , Hibridomas/imunologia , Lectinas Tipo C/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Masculino , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos , Receptores de Superfície Celular/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
11.
Haemophilia ; 19(3): 399-402, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23252683

RESUMO

The administration of therapeutic factor VIII (FVIII) to patients with haemophilia A induces the development of inhibitory anti-FVIII IgG in a substantial number of patients. For an antigen-specific immune response to develop, antigen-presenting cells (APCs) need to mature and procure appropriate co-stimulatory signals to T cells at the time of presentation of the endocytosed antigen. The nature of the danger signals that induce APC maturation, thus initiating the anti-FVIII immune response, are yet ill-characterized. Contradictory reports on a direct effect of therapeutic FVIII on APC maturation have been released. Here, we investigated whether FVIII directly triggers Toll-like receptor 2 (TLR2) signalling. The capacity of human recombinant FVIII to promote the maturation of a mouse bone marrow macrophage cell line (BMA) was investigated by flow cytometry. In parallel, the triggering of TLR1.2 or TLR2.6-expressing HEK293 cells by FVIII was analysed following transfection of the cells with a reporter construct for NFκB activity. In contrast, to zymosan, a known TLR2 agonist, human recombinant FVIII did not induce the maturation of mouse BMA macrophages, as analysed by the levels of expression of CD80, CD86, CD40 and I-Ab at the cell surface. Furthermore, incubation of FVIII with cells expressing TLR2 paired with TLR1 or TLR6, failed to activate NFκB, whereas NKκB activity was triggered in the presence of zymosan. Our results confirm that FVIII alone is insufficient to trigger the maturation of APCs that is required to initiate an immune response.


Assuntos
Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Receptor 1 Toll-Like/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 6 Toll-Like/metabolismo , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos de Superfície/metabolismo , Células da Medula Óssea/citologia , Linhagem Celular , Fator VIII/farmacologia , Células HEK293 , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 1 Toll-Like/genética , Receptor 2 Toll-Like/agonistas , Receptor 2 Toll-Like/genética , Receptor 6 Toll-Like/genética , Transfecção , Zimosan/farmacologia
12.
J Thromb Haemost ; 10(6): 1062-71, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22471307

RESUMO

BACKGROUND: Heme is a redox active macrocyclic compound that is released upon tissue damage or hemorrhages. The extracellular release of large amounts of heme saturates scavenging heme-binding proteins. Free heme has been proposed to affect coagulation and has been co-purified with the factor VIII (FVIII)-von Willebrand factor (VWF) complex. The sites from which heme is released upon injury overlap with the sites to which FVIII is targeted for performing its hemostatic functions. OBJECTIVES: To investigate the interaction of heme with FVIII and the consequence for the procoagulant activity of FVIII in vitro. METHODS AND RESULTS: Heme bound to several sites on FVIII with high apparent affinity. Heme-binding inhibited FVIII procoagulant activity in a dose-dependent manner. FVIII inactivation in the presence of saturating amounts of heme implicated a reduced interaction of FVIII with activated FIX, as shown by ELISA, surface plasmon resonance and fluorescence quenching. Heme-mediated inactivation of FVIII was prevented by VWF, but not by human serum albumin, a heme-binding protein known for its protective activity in hemolytic conditions. CONCLUSIONS: Our data identify FVIII as a novel heme-binding protein. Occupation of high affinity heme-binding sites on FVIII at low concentrations of free heme did not inactivate FVIII. Conversely, large molar excesses of heme over FVIII, which correspond to conditions of extensive heme release, inhibited FVIII activity in vitro. It remains to be demonstrated whether, under such conditions, heme-mediated modulation of the activity of FVIII plays some role in the regulation of coagulation.


Assuntos
Coagulação Sanguínea , Fator IXa/metabolismo , Fator VIII/metabolismo , Heme/metabolismo , Sítios de Ligação , Ligação Competitiva , Ensaio de Imunoadsorção Enzimática , Fator VIIIa/metabolismo , Hemina/metabolismo , Humanos , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Proteínas Recombinantes/metabolismo , Espectrometria de Fluorescência , Ressonância de Plasmônio de Superfície , Fator de von Willebrand/metabolismo
13.
Haemophilia ; 18(2): 248-54, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22044692

RESUMO

The development of inhibitory anti-factor VIII (FVIII) antibodies in patients with haemophilia A following replacement therapy is associated with several types of risk factors. Among these, the purity of FVIII concentrates, and in particular the presence of von Willebrand factor (VWF), was controversially proposed to influence the immunogenicity of exogenous FVIII. We re-assessed in vivo and in vitro the immuno-protective effect of VWF towards FVIII. The immuno-protective effect of VWF towards FVIII was investigated in vivo, in a model of haemophilia A. We studied the endocytosis of FVIII by murine bone marrow-derived dendritic cells and evaluated the capacity of VWF to block the internalization of FVIII. We characterized the relevance of VWF for the accumulation of FVIII in the marginal zone of the spleen, a secondary lymphoid organ where the immune response to therapeutically administered FVIII initiates. Our results confirm that VWF reduces the immunogenicity of FVIII in FVIII-deficient mice. Paradoxically, VWF is important for the accumulation of FVIII in the marginal zone of the spleen. We propose that VWF exerts at least two non-mutually exclusive immunoprotective roles towards FVIII in haemophilic mice: VWF prevents the endocytosis of FVIII by professional antigen-presenting cells by blocking the interaction of FVIII with as yet unidentified endocytic receptor(s). Hypothetically, VWF, by virtue of increasing the half-life of FVIII in the circulation, may allow an increased contact time with tolerogenic marginal zone B cells in the spleen.


Assuntos
Fator VIII/imunologia , Hemofilia A/imunologia , Fator de von Willebrand/fisiologia , Animais , Células Dendríticas/efeitos dos fármacos , Modelos Animais de Doenças , Endocitose/imunologia , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL
14.
Clin Exp Immunol ; 164 Suppl 2: 2-5, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21466545

RESUMO

Intravenous immunoglobulin (IVIG) is a therapeutic compound prepared from pools of plasma obtained from several thousand healthy blood donors. For more than 20 years, IVIG has been used in the treatment of a wide range of primary and secondary immunodeficiencies. IVIG now represents a standard therapeutic option for most antibody deficiencies. Routinely, IVIG is used in patients with X-linked agammaglobulinaemia (XLA), common variable immunodeficiency (CVID), X-linked hyper-IgM, severe combined immunodeficiency, Wiskott-Aldrich syndrome, and selective IgG class deficiency. In addition, IVIG is used extensively in the treatment of a wide variety of autoimmune disorders. IVIG is administered at distinct doses in the two clinical settings: whereas immunodeficient patients are treated with replacement levels of IVIG, patients with autoimmune and inflammatory diseases are administered with very high doses of IVIG. Several lines of experimental evidence gathered in the recent years suggest that the therapeutic beneficial effect of IVIG in immunodeficiencies reflects an active role for IVIG, rather than a mere passive transfer of antibodies.


Assuntos
Imunoglobulinas Intravenosas/uso terapêutico , Síndromes de Imunodeficiência/tratamento farmacológico , Doenças Autoimunes/tratamento farmacológico , Humanos , Resultado do Tratamento
15.
J Thromb Haemost ; 9(4): 719-28, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21251202

RESUMO

BACKGROUND: Replacement therapy with exogenous factor VIII to treat hemorrhages induces inhibitory anti-FVIII antibodies in up to 30% of patients with hemophilia A. Current approaches to eradicate FVIII inhibitors using high-dose FVIII injection protocols (immune tolerance induction) or anti-CD20 depleting antibodies (Rituximab) demonstrate limited efficacy; they are extremely expensive and/or require stringent compliance from the patients. OBJECTIVES: To investigate whether the proteasome inhibitor bortezomib, which depletes plasmocytes, modulates the anti-FVIII immune response in FVIII-deficient mice. METHODS AND RESULTS: Preventive 4-week treatment of naïve mice with bortezomib at the time of FVIII administration delayed the development of inhibitory anti-FVIII IgG, and depleted plasma cells as well as different lymphoid cell subsets. Conversely, curative treatment of inhibitor-positive mice for 10 weeks, along with FVIII administration, failed to eradicate FVIII inhibitors to extents that would be clinically relevant if achieved in patients. Accordingly, bortezomib did not eradicate anti-FVIII IgG-secreting plasmocytes that had homed to survival niches in the bone marrow, despite significant elimination of total plasma cells. CONCLUSIONS: The data suggest that strategies for the efficient reduction of anti-FVIII IgG titers in patients with hemophilia A should rely on competition for survival niches for plasmocytes in the bone marrow rather than the mere use of proteasome inhibitors.


Assuntos
Ácidos Borônicos/uso terapêutico , Fator VIII/antagonistas & inibidores , Imunoglobulina G/imunologia , Pirazinas/uso terapêutico , Inibidores de Serina Proteinase/uso terapêutico , Animais , Bortezomib , Ensaio de Imunoadsorção Enzimática , Fator VIII/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
16.
J Thromb Haemost ; 8(7): 1524-31, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20374449

RESUMO

SUMMARY BACKGROUND: BO2C11 is a human monoclonal factor (F) VIII inhibitor. When bound to the C2 domain of FVIII, the Fab fragment of BO2C11 (Fab(BO2C11)) buries a surface of C2 that contains residues participating in a binding site for von Willebrand factor (VWF). BO2C11 has thus been proposed to neutralize FVIII by steric hindrance. OBJECTIVES: The BO2C11 epitope on C2 overlaps with residues located at the periphery of the putative VWF binding site; hence, most of the residues that constitute the VWF binding site on C2 and a3 remain accessible for VWF interaction following BO2C11/FVIII complex formation. We thus investigated the contribution of alternative molecular mechanisms to FVIII inactivation by BO2C11. METHODS: Continuum electrostatic calculations were applied to the crystal structure of C2, free or Fab(BO2C11)-complexed. In silico predictions were confirmed by site-directed mutagenesis and VWF-binding assays of the mutated FVIII. RESULTS: Binding of Fab(BO2C11) to C2 induced perturbations in the electrostatic potential of C2 and in the local electrostatic parameters of 18 charged residues in C2, which are distant from the BO2C11 epitope. Nine of the predicted electrostatic hotspots clustered on the VWF-binding site of C2. Mutation of some of the predicted electrostatic hotspots has been associated with hemophilia A and reduced VWF binding in vitro. CONCLUSIONS: Inhibitors may neutralize FVIII by alteration of protein surface electrostatics at a long distance from their epitope. Perturbation of the electrostatic environment of C2, either upon binding by anti-FVIII antibodies or consecutive to missense mutations in the F8 gene, may lead to hampered VWF binding and reduced FVIII residence time in circulation.


Assuntos
Anticorpos Monoclonais/farmacologia , Epitopos/imunologia , Fator VIII/imunologia , Eletricidade Estática , Fator de von Willebrand/metabolismo , Anticorpos Monoclonais/imunologia , Sítios de Ligação/efeitos dos fármacos , Fator VIII/antagonistas & inibidores , Fator VIII/química , Fator VIII/genética , Hemofilia A , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto
17.
J Thromb Haemost ; 7(12): 2006-2015, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19817985

RESUMO

BACKGROUND: Approximately 25% of severe hemophilia A (HA) patients develop antibodies to factor VIII protein. PATIENTS: In the present case-controlled cohort study, 260 severely affected, mutation-type-matched HA patients were studied for association of human leukocyte antigen (HLA) class II molecules and polymorphisms in the genes encoding interleukin-10 (IL-10), tumor necrosis factor-alpha (TNF-alpha) and cytotoxic T-lymphocyte antigen-4 (CTLA-4) and development of inhibitors. RESULTS: Our results demonstrate a higher frequency of DRB1*15 and DQB1*0602 alleles as well as of the haplotype DRB1*15/DQB1*0602 in inhibitor patients [odds ratio (OR) 1.9; P < 0.05]. In TNF-alp[ha, the A allele of the 308G>A polymorphism was found with higher frequency in the inhibitor cohort (0.22 vs. 0.13, OR 1.80). This finding was more pronounced for the homozygous A/A genotype (OR 4.7). For IL-10, the 1082G allele was observed more frequently in patients with inhibitors (0.55 vs. 0.43; P = 0.008). The functional cytokine phenotype was determined for the first time, on the basis of the genetic background, and this showed that 12% of patients with inhibitors were high-TNF-alpha/high-IL-10 producers, as compared with 3% of non-inhibitor patients (OR 4.4). A trend for a lower frequency of the A allele of the CT60 polymorphism in CTLA-4 was found in inhibitor patients (0.42 vs. 0.50). CONCLUSIONS: In conclusion, the reported data clearly highlighted the participation of HLA molecules in inhibitor formation in a large cohort of patients. The higher frequencies of the 308G>A polymorphism in TNF-alpha and 1082A>G in IL-10 in inhibitor patients confirmed the earlier published data. The CT60 single-nucleotide polymorphism in CTLA-4 is of apparently less importance.


Assuntos
Antígenos CD/genética , Autoanticorpos/biossíntese , Genes MHC da Classe II/genética , Hemofilia A/imunologia , Interleucina-10/genética , Polimorfismo Genético/imunologia , Fator de Necrose Tumoral alfa/genética , Autoanticorpos/genética , Antígeno CTLA-4 , Frequência do Gene , Antígenos HLA-DQ/genética , Cadeias beta de HLA-DQ , Antígenos HLA-DR/genética , Cadeias HLA-DRB1 , Hemofilia A/epidemiologia , Hemofilia A/genética , Humanos
18.
J Thromb Haemost ; 7(11): 1816-23, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19682235

RESUMO

BACKGROUND: Alloimmune responses to intravenously administered protein therapeutics are the most common cause of failure of replacement therapy in patients with defective levels of endogenous proteins. Such a situation is encountered in some patients with hemophilia A, who develop inhibitory anti-factor (F)VIII alloantibodies after administration of FVIII to treat hemorrhages. OBJECTIVES: The nature of the secondary lymphoid organs involved in the initiation of immune responses to human therapeutic has not been studied. We therefore investigated this in the case of FVIII, a self-derived exogenous protein therapeutic. METHODS: The distribution of intravenously administered FVIII was followed after FVIII-deficient mice were injected with radiolabeled FVIII and using immunohistochemistry. The role of the spleen and antigen-presenting cells (APC) in the onset of the anti-FVIII immune response was analyzed upon splenectomy or treatment of the mice with APC-depleting compounds. RESULTS: FVIII preferentially accumulated in the spleen at the level of metallophilic macrophages in the marginal zone (MZ). Surgical removal of the spleen or selective in vivo depletion of macrophages and CD11c-positive CD8 alpha-negative dendritic cells resulted in a drastic reduction in anti-FVIII immune responses. CONCLUSIONS: Using FVIII-deficient mice as a model for patients with hemophilia A, and human pro-coagulant FVIII as a model for immunogenic self-derived protein therapeutics, our results highlight the importance of the spleen and MZ APCs in the initiation of immune responses to protein therapeutics. Identification of the receptors implicated in retention of protein therapeutics in the MZ may pave the way towards novel strategies aimed at reducing their immunogenicity.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Fator VIII/farmacocinética , Hemofilia A/imunologia , Isoanticorpos/sangue , Baço/imunologia , Animais , Fator VIII/imunologia , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Imunidade/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Baço/citologia , Esplenectomia , Distribuição Tecidual
19.
Haemophilia ; 14 Suppl 3: 36-42, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18510520

RESUMO

The immune response to factor VIII and the development of inhibitory antibodies is a complex multi-factorial process involving a variety of immune regulatory genes and cells, several of which have the potential to determine risk. A better understanding of the mechanisms involved will increase the likelihood of development of new therapeutic options for patients with hemophilia. This review summarizes genetic and non-genetic risk factors currently under evaluation, and the potential modulative effect of the von Willebrand factor on factor VIII immuno- and antigenicity. In addition, the role of T-regulatory cells in the pathogenicity of inhibitors will be discussed.


Assuntos
Inibidores dos Fatores de Coagulação Sanguínea/imunologia , Fator VIII/imunologia , Hemofilia A/genética , Polimorfismo Genético/genética , Autoanticorpos/imunologia , Autoanticorpos/metabolismo , Autoimunidade/imunologia , Terapia Genética/métodos , Hemofilia A/imunologia , Humanos , Polimorfismo Genético/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Fator de von Willebrand/imunologia
20.
Haemophilia ; 13 Suppl 5: 61-4, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18078399

RESUMO

In patients with haemophilia A, factor VIII (FVIII) therapy leads to the development of anti-FVIII alloantibodies that inhibit FVIII pro-coagulant activity, in up to 25% of the cases. At a time when efficient viral screening procedures are at place, development of inhibitors poses the greatest threat to haemophilia A patients. Various risk factors, both patient and product-related, are responsible for the development of inhibitory antibodies. The role of FVIII-specific CD4+ T lymphocytes in the initiation of the humoral immune response to exogenous FVIII has been well. In view of their capacity to stimulate naïve T cells, dendritic cells (DCs) play a central role in the initiation of the primary immune response. Thus, in the context of a primary alloimmunization against FVIII, i.e. when FVIII-specific B lymphocytes are not there to take up FVIII from the circulation and to serve as antigen presenting cells (APCs), DCs are the only cell type that internalize FVIII, leading to activation of FVIII-specific CD4+ T lymphocytes. von Willebrand factor (VWF) present in plasma-derived FVIII therapeutic concentrates, is known to act as a chaperone molecule for procoagulant FVIII. In addition to its role in reducing the 'antigenicity' of FVIII, the role of VWF in the reduction of the 'immunogenicity' of therapeutic FVIII in patients with haemophilia A has also been suggested. We have recently demonstrated that VWF protects FVIII from being endocytosed by human DCs and subsequently being presented to FVIII-specific T cells. We propose that VWF may reduce the immunogenicity of FVIII by preventing, upstream from the activation of immune effectors, the entry of FVIII in professional antigen presenting cells.


Assuntos
Coagulantes/imunologia , Células Dendríticas/imunologia , Fator VIII/imunologia , Hemofilia A/imunologia , Isoanticorpos/imunologia , Fator de von Willebrand/fisiologia , Apresentação de Antígeno , Linfócitos T CD4-Positivos/imunologia , Coagulantes/uso terapêutico , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Humanos , Tolerância Imunológica , Ativação Linfocitária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...