Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
3.
Acta Naturae ; 7(2): 74-9, 2015.
Article in English | MEDLINE | ID: mdl-26085947

ABSTRACT

B cells play a crucial role in the development and pathogenesis of systemic and organ-specific autoimmune diseases. Autoreactive B cells not only produce antibodies, but also secrete pro-inflammatory cytokines and present specific autoantigens to T cells. The treatment of autoimmune diseases via the elimination of the majority of B cells using the monoclonal anti-CD19/20 antibody (Rituximab) causes systemic side effects and, thus, requires a major revision. Therapeutic intervention directed towards selective elimination of pathogenic autoreactive B cells has the potential to become a universal approach to the treatment of various autoimmune abnormalities. Here, we developed a recombinant immunotoxin based on the immunodominant peptide of the myelin basic protein (MBP), fused to the antibody Fc domain. We showed that the obtained immunotoxin provides selective in vivo elimination of autoreactive B cells in mice with experimental autoimmune encephalomyelitis. The proposed conception may be further used for the development of new therapeutics for a targeted treatment of multiple sclerosis and other autoimmune disorders.

6.
J Thromb Haemost ; 12(12): 2065-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25267332

ABSTRACT

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.


Subject(s)
Factor VIII/immunology , Hemophilia A/blood , Immunity, Humoral , Animals , Antibodies, Neutralizing/immunology , Blood Coagulation , Disease Models, Animal , Hemophilia A/genetics , Inflammation , Mice , Mutation , Plasmids , Protein Structure, Tertiary , Recombinant Proteins/immunology , Thrombin/chemistry , Thromboplastin/chemistry , Warfarin/pharmacology
7.
Haemophilia ; 20(2): 176-84, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24118514

ABSTRACT

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.


Subject(s)
Factor VIII/genetics , Factor VIII/immunology , Hemophilia A/genetics , Hemophilia A/immunology , Histocompatibility Antigens Class II/genetics , Isoantibodies/immunology , Mutation, Missense , Peptide Fragments/immunology , Amino Acid Sequence , Databases, Genetic , Epitopes/chemistry , Epitopes/immunology , Factor VIII/chemistry , Hemophilia A/drug therapy , Histocompatibility Antigens Class II/immunology , Humans , Protein Binding/immunology , Risk Factors
8.
Haemophilia ; 20(4): 486-92, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24372710

ABSTRACT

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.


Subject(s)
Factor VIII/adverse effects , Factor VIII/immunology , Animals , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endocytosis , Factor VIII/metabolism , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Hemophilia A/immunology , Humans , Hybridomas/immunology , Lectins, C-Type/metabolism , Lymphocyte Activation/drug effects , Male , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Receptors, Cell Surface/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
9.
Haemophilia ; 19(3): 399-402, 2013 May.
Article in English | MEDLINE | ID: mdl-23252683

ABSTRACT

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.


Subject(s)
Factor VIII/therapeutic use , Hemophilia A/drug therapy , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 6/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, Surface/metabolism , Bone Marrow Cells/cytology , Cell Line , Factor VIII/pharmacology , HEK293 Cells , Humans , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 1/genetics , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/genetics , Toll-Like Receptor 6/genetics , Transfection , Zymosan/pharmacology
10.
J Thromb Haemost ; 10(6): 1062-71, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22471307

ABSTRACT

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.


Subject(s)
Blood Coagulation , Factor IXa/metabolism , Factor VIII/metabolism , Heme/metabolism , Binding Sites , Binding, Competitive , Enzyme-Linked Immunosorbent Assay , Factor VIIIa/metabolism , Hemin/metabolism , Humans , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Recombinant Proteins/metabolism , Spectrometry, Fluorescence , Surface Plasmon Resonance , von Willebrand Factor/metabolism
11.
Haemophilia ; 18(2): 248-54, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22044692

ABSTRACT

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.


Subject(s)
Factor VIII/immunology , Hemophilia A/immunology , von Willebrand Factor/physiology , Animals , Dendritic Cells/drug effects , Disease Models, Animal , Endocytosis/immunology , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Male , Mice , Mice, Inbred C57BL
12.
Clin Exp Immunol ; 164 Suppl 2: 2-5, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21466545

ABSTRACT

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.


Subject(s)
Immunoglobulins, Intravenous/therapeutic use , Immunologic Deficiency Syndromes/drug therapy , Autoimmune Diseases/drug therapy , Humans , Treatment Outcome
13.
J Thromb Haemost ; 9(4): 719-28, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21251202

ABSTRACT

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.


Subject(s)
Boronic Acids/therapeutic use , Factor VIII/antagonists & inhibitors , Immunoglobulin G/immunology , Pyrazines/therapeutic use , Serine Proteinase Inhibitors/therapeutic use , Animals , Bortezomib , Enzyme-Linked Immunosorbent Assay , Factor VIII/immunology , Male , Mice , Mice, Inbred C57BL
14.
Parasite Immunol ; 32(6): 464-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20500678

ABSTRACT

Chagas disease induced by Trypanosoma cruzi (Tc) infection is an important cause of mortality and morbidity affecting the cardiovascular system for which presently available therapies are insufficient and largely inadequate. Intravenous immunoglobulin (IVIg) is a therapeutic preparation containing normal polyspecific IgG obtained from plasma pools of several thousand healthy donors and is used in several autoimmune, inflammatory and infectious diseases. In the study of heart from mice chronically infected with Tc, we observed that IVIg restores type 1 atrioventricular block or bradycardia. In the present study, we investigated the effects of IVIg in acute Tc infection. Intravenous immunoglobulin administration after the first week of infection was associated with an increase in survival time. Taken together, results observed in the chronic and in the acute phase associate IVIg treatment with a favourable outcome in T. cruzi infection.


Subject(s)
Chagas Disease/therapy , Immunoglobulins, Intravenous/therapeutic use , Animals , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred BALB C , Survival Analysis , Time Factors
15.
J Thromb Haemost ; 8(7): 1524-31, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20374449

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/pharmacology , Epitopes/immunology , Factor VIII/immunology , Static Electricity , von Willebrand Factor/metabolism , Antibodies, Monoclonal/immunology , Binding Sites/drug effects , Factor VIII/antagonists & inhibitors , Factor VIII/chemistry , Factor VIII/genetics , Hemophilia A , Humans , Models, Molecular , Mutagenesis, Site-Directed , Mutation, Missense
16.
Rev Med Interne ; 30(12 Suppl): H1-8, 2009 Dec.
Article in French | MEDLINE | ID: mdl-19995650

ABSTRACT

The recognition of self is a normal function of the immune system. A dysregulation of the tight control of auto-reactive lymphocytes in physiological conditions sometimes leads to the development of auto-immune diseases. Several recent elements bring new insights in the functioning of the immune system. Thus, the discovery of BAFF and APRIL and their receptors allow us to better understand the homeostasis and activation of B lymphocytes. The description of a new helper lineage, characterized by the secretion of IL-17 modifies the etiologic scheme of auto-immune diseases. Lastly, regulatory T lymphocytes play a major role in controlling auto-reactive lymphocytes and may participate in the genesis of auto-immune diseases. The emergence of these new data enables us to better understand the pathological mechanisms of complex auto-immunes diseases. However, further studies are necessary in order to specify the role of each one of these factors. This will enable a better targeting of treatments in order to improve the management of patients.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity/physiology , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Humans , Immunity, Innate/physiology
17.
Clin Exp Immunol ; 158 Suppl 1: 2-13, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19883419

ABSTRACT

High-dose intravenous immunoglobulin (IVIg) preparations are used currently for the treatment of autoimmune or inflammatory diseases. Despite numerous studies demonstrating efficacy, the precise mode of action of IVIg remains unclear. Paradoxically, IgG can exert both pro- and anti-inflammatory activities, depending on its concentration. The proinflammatory activity of low-dose IVIg requires complement activation or binding of the Fc fragment of IgG to IgG-specific receptors (FcgammaR) on innate immune effector cells. In contrast, when administered in high concentrations, IVIg has anti-inflammatory properties. How this anti-inflammatory effect is mediated has not yet been elucidated fully, and several mutually non-exclusive mechanisms have been proposed. This paper represents the proceedings of a session entitled 'IVIg--Understanding properties and mechanisms' at the 6th International Immunoglobulin Symposium that was held in Interlaken on 26-28 March 2009. The presentations addressed how IgG may affect the cellular compartment, evidence for IVIg-mediated scavenging of complement fragments, the role of the dimeric fraction of IVIg, the anti-inflammatory properties of the minor fraction of sialylated IgG molecules, and the genetic organization and variation in FcgammaRs. These findings demonstrate the considerable progress that has been made in understanding the mechanisms of action of IVIgs, and may influence future perspectives in the field of Ig therapy.


Subject(s)
Immunoglobulins, Intravenous/therapeutic use , Animals , Dendritic Cells/immunology , Disease Models, Animal , Humans , Immunoglobulin G/immunology , Immunoglobulin G/therapeutic use , Immunoglobulins, Intravenous/immunology , Immunomodulation/immunology , Inflammation/therapy , Mice , Polymorphism, Single Nucleotide , Receptors, IgG/genetics , Receptors, IgG/immunology
18.
Clin Exp Immunol ; 158 Suppl 1: 60-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19883425

ABSTRACT

The posters presented at the 6th International Immunoglobulin Symposium covered a wide range of fields and included both basic science and clinical research. From the abstracts accepted for poster presentation, 12 abstracts were selected for oral presentations in three parallel sessions on immunodeficiencies, autoimmunity and basic research. The immunodeficiency presentations dealt with novel, rare class-switch recombination (CSR) deficiencies, attenuation of adverse events following IVIg treatment, association of immunoglobulin (Ig)G trough levels and protection against acute infection in patients with X-linked agammaglobulinaemia (XLA) and common variable immunodeficiency (CVID), and the reduction of class-switched memory B cells in patients with specific antibody deficiency (SAD). The impact of intravenous immunoglobulin on fetal alloimmune thrombocytopenia, pregnancy and postpartum-related relapses in multiple sclerosis and refractory myositis, as well as experiences with subcutaneous immunoglobulin in patients with multi-focal motor neuropathy, were the topics presented in the autoimmunity session. The interaction of dendritic cell (DC)-SIGN and alpha2,6-sialylated IgG Fc and its impact on human DCs, the enrichment of sialylated IgG in plasma-derived IgG, as wells as prion surveillance and monitoring of anti-measles titres in immunoglobulin products, were covered in the basic science session. In summary, the presentations illustrated the breadth of immunoglobulin therapy usage and highlighted the progress that is being made in diverse areas of basic and clinical research, extending our understanding of the mechanisms of immunoglobulin action and contributing to improved patient care.


Subject(s)
Immunoglobulins/therapeutic use , Immunologic Deficiency Syndromes/drug therapy , Autoimmune Diseases/drug therapy , Autoimmunity/immunology , Biomedical Research , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunologic Deficiency Syndromes/immunology
20.
J Thromb Haemost ; 7(11): 1816-23, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19682235

ABSTRACT

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.


Subject(s)
Antigen-Presenting Cells/immunology , Factor VIII/pharmacokinetics , Hemophilia A/immunology , Isoantibodies/blood , Spleen/immunology , Animals , Factor VIII/immunology , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Immunity/drug effects , Macrophages/immunology , Mice , Spleen/cytology , Splenectomy , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL