Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 44
Filtrer
1.
Front Immunol ; 11: 515556, 2020.
Article de Anglais | MEDLINE | ID: mdl-33178180

RÉSUMÉ

Highly sensitive reporter-gene assays have been developed that allow both the direct vascular endothelial growth factor (VEGF) neutralizing activity of bevacizumab and the ability of bevacizumab to activate antibody dependent cellular cytotoxicity (ADCC) to be quantified rapidly and in a highly specific manner. The use of these assays has shown that in 46 patients with ovarian cancer following four cycle of bevacizumab treatment, and in longitudinal samples from the two patients that respond to bevacizumab therapy from a small cohort of patients with glioblastoma, that there is a reasonably good correlation between bevacizumab drug levels determined by ELISA and bevacizumab activity, determined using either the VEGF-responsive reporter gene, or the ADCC assays. One of the two primary non-responders with glioblastoma exhibited high levels of ADCC activity suggesting reduced bevacizumab Fc engagement in vivo in contrast to the other primary non-responder, and the two secondary non-responders with a decreasing bevacizumab PK profile, determined by ELISA that exhibited low to undetectable ADCC activity. Drug levels were consistently higher than bevacizumab activity determined using the reporter gene assay in serial samples from one of the secondary non-responders and lower in some samples from the other secondary non-responder and ADCC activity was markedly lower in all samples from these patients suggesting that bevacizumab activity may be partially neutralized by anti-drug neutralizing antibodies (NAbs). These results suggest that ADCC activity may be correlated with the ability of some patients to respond to treatment with bevacizumab while the use of the VEGF-responsive reporter-gene assay may allow the appearance of anti-bevacizumab NAbs to be used as a surrogate maker of treatment failure prior to the clinical signs of disease progression.


Sujet(s)
Bévacizumab/administration et posologie , Glioblastome , Protéines tumorales/immunologie , Tumeurs de l'ovaire , Facteur de croissance endothéliale vasculaire de type A/immunologie , Lignée cellulaire tumorale , Femelle , Glioblastome/traitement médicamenteux , Glioblastome/immunologie , Glioblastome/anatomopathologie , Cellules HEK293 , Humains , Études longitudinales , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/anatomopathologie
2.
J Immunol Res ; 2017: 3908289, 2017.
Article de Anglais | MEDLINE | ID: mdl-29104875

RÉSUMÉ

Novel ADCC effector cells expressing the V-variant or F-variant of FcγRIIIa (CD16a) and firefly luciferase under the control of a chimeric promoter incorporating recognition sequences for the principal transcription factors involved in FcγRIIIa signal transduction, together with novel target cells overexpressing a constant high level of the specific antigen recognized by rituximab, trastuzumab, cetuximab, infliximab, adalimumab, or etanercept, confer improved sensitivity, specificity, and dynamic range in an ADCC assay relative to effector cells expressing a NFAT-regulated reporter gene and wild-type target cells. The effector cells also contain a normalization gene rendering ADCC assays independent of cell number or serum matrix effects. The novel effector and target cells in a frozen thaw-and-use format exhibit low vial-to-vial and lot-to-lot variation in their performance characteristics reflected by CVs of 10% or less. Homologous control target cells in which the specific target gene has been invalidated by genome editing providing an ideal control and a means of correcting for nonspecific effects were observed with certain samples of human serum. The novel effector cells and target cells expressing noncleavable membrane-bound TNFα have been used to quantify ADCC activity in serum from patients with Crohn's disease treated with infliximab and to relate ADCC activity to drug levels.


Sujet(s)
Cytotoxicité à médiation cellulaire dépendante des anticorps , Antigènes CD20/génétique , Maladie de Crohn/immunologie , Récepteurs ErbB/génétique , Techniques immunologiques/méthodes , Facteurs de transcription NFATC/génétique , Récepteur ErbB-2/génétique , Récepteurs du fragment Fc des IgG/génétique , Lymphocytes T/physiologie , Facteur de nécrose tumorale alpha/génétique , Antigènes CD20/immunologie , Cétuximab/métabolisme , Récepteurs ErbB/immunologie , Étanercept/métabolisme , Gènes rapporteurs/génétique , Cellules HEK293 , Humains , Infliximab/métabolisme , Cellules Jurkat , Récepteur ErbB-2/immunologie , Récepteurs du fragment Fc des IgG/immunologie , Rituximab/métabolisme , Transduction du signal , Transgènes/génétique , Trastuzumab/métabolisme , Facteur de nécrose tumorale alpha/immunologie
3.
Eur Cytokine Netw ; 27(3): 63-67, 2016 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-27910810

RÉSUMÉ

OBJECTIVE: To investigate the relationship between the response to influenza vaccination and the ability to produce proinflamatory cytokines in elderly subjects. METHODS: Whole blood samples from 25 elderly subjects collected before influenza vaccination were stimulated with the influenza vaccine in order to evaluate the secretion of five specific cytokines: TNFα, IFNα, IFNγ, IL2 and IL10. The results were correlated with the increased HAI antibody titres two weeks after vaccination. RESULTS: Only 30% of elderly individuals seroconverted after vaccination. Although 50 to 70% of the cohort did not produce TNFα, IFNα, IFNγ, IL2 or IL10, all of the individuals who seroconverted were able to produce TNFα. Furthermore production of IFNγ, with or without production of IFNα/ß, was not associated with a better response to the vaccine. CONCLUSION: Production of TNFα appears to be primordial for an efficient vaccine response, and may provide a predictive marker for the humoral response to vaccination. It may also provide the basis for evaluating agents designed to rescue TNFα-producing cells. This study emphasises a need to rescue TNF-producing cell function.


Sujet(s)
Virus de la grippe A/immunologie , Vaccins antigrippaux/immunologie , Grippe humaine/sang , Grippe humaine/prévention et contrôle , Facteur de nécrose tumorale alpha/biosynthèse , Sujet âgé , Sujet âgé de 80 ans ou plus , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Cellules cultivées , Cytokines/biosynthèse , Femelle , Humains , Virus de la grippe A/classification , Grippe humaine/immunologie , Grippe humaine/virologie , Mâle , Vaccination
4.
J Leukoc Biol ; 99(1): 175-88, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-26232452

RÉSUMÉ

Although interferon-ß is used as first-line therapy for multiple sclerosis, the cell type-specific activity of type I interferons in multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis, remains obscure. In this study, we have elucidated the in vivo immunomodulatory role of type I interferon signaling in T cells during experimental autoimmune encephalomyelitis by use of a novel transgenic mouse, carrying a cd2-ifnar1 transgene on a interferon-α/ß receptor 1 null genetic background, thus allowing expression of the interferon-α/ß receptor 1 and hence, a functional type I interferon receptor exclusively on T cells. These transgenic mice exhibited milder experimental autoimmune encephalomyelitis with reduced T cell infiltration, demyelination, and axonal damage in the central nervous system. It is noteworthy that interferon-ß administration in transgenic mice generated a more pronounced, protective effect against experimental autoimmune encephalomyelitis compared with untreated littermates. In vivo studies demonstrated that before experimental autoimmune encephalomyelitis onset, endogenous type I interferon receptor signaling in T cells led to impaired T-helper 17 responses, with a reduced fraction of CCR6(+) CD4(+) T cells in the periphery. At the acute phase, an increased proportion of interleukin-10- and interferon-γ-producing CD4(+) T cells was detected in the periphery of the transgenic mice, accompanied by up-regulation of the interferon-γ-induced gene Irgm1 in peripheral T cells. Together, these results reveal a hitherto unknown T cell-associated protective role of type I interferon in experimental autoimmune encephalomyelitis that may provide valuable clues for designing novel therapeutic strategies for multiple sclerosis.


Sujet(s)
Activation des lymphocytes/immunologie , Récepteur à l'interféron alpha-bêta/métabolisme , Transduction du signal , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Animaux , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Système nerveux central/immunologie , Système nerveux central/métabolisme , Système nerveux central/anatomopathologie , Analyse de regroupements , Cytokines/métabolisme , Modèles animaux de maladie humaine , Encéphalomyélite auto-immune expérimentale , Expression des gènes , Analyse de profil d'expression de gènes , Interféron de type I/métabolisme , Souris , Souris knockout , Souris transgéniques , Glycoprotéine MOG/immunologie , Spécificité d'organe/génétique , Fragments peptidiques/immunologie , Récepteur à l'interféron alpha-bêta/génétique
5.
J Virol ; 89(5): 2731-8, 2015 Mar.
Article de Anglais | MEDLINE | ID: mdl-25540366

RÉSUMÉ

UNLABELLED: Previously we found that following intranasal (i.n.) infection with neurotropic vesicular stomatitis virus (VSV) type I interferon receptor (IFNAR) triggering of neuroectodermal cells was critically required to constrain intracerebral virus spread. To address whether locally active IFN-ß was induced proximally, we studied spatiotemporal conditions of VSV-mediated IFN-ß induction. To this end, we performed infection studies with IFN-ß reporter mice. One day after intravenous (i.v.) VSV infection, luciferase induction was detected in lymph nodes. Upon i.n. infection, luciferase induction was discovered at similar sites with delayed kinetics, whereas on days 3 and 4 postinfection enhanced luciferase expression additionally was detected in the foreheads of reporter mice. A detailed analysis of cell type-specific IFN-ß reporter mice revealed that within the olfactory bulb IFN-ß was expressed by neuroectodermal cells, primarily by astrocytes and to a lesser extent by neurons. Importantly, locally induced type I IFN triggered distal parts of the brain as indicated by the analysis of ISRE-eGFP mice which after i.n. VSV infection showed enhanced green fluorescent protein (eGFP) expression throughout the brain. Compared to wild-type mice, IFN-ß(-/-) mice showed increased mortality to i.n. VSV infection, whereas upon i.v. infection no such differences were detected highlighting the biological significance of intracerebrally expressed IFN-ß. In conclusion, upon i.n. VSV instillation, IFN-ß responses mounted by astrocytes within the olfactory bulb critically contribute to the antiviral defense by stimulating distal IFN-ß-negative brain areas and thus arresting virus spread. IMPORTANCE: The central nervous system has long been considered an immune privileged site. More recently, it became evident that specialized immune mechanisms are active within the brain to control pathogens. Previously, we showed that virus, which entered the brain via the olfactory route, was arrested within the olfactory bulb by a type I IFN-dependent mechanism. Since peripheral type I IFN would not readily cross the blood-brain barrier and within the brain thus far no abundant type I IFN responses have been detected, here we addressed from where locally active IFN originated from. We found that upon intranasal VSV instillation, primarily astrocytes, and to a lesser extent neurons, were stimulated within the olfactory bulb to mount IFN-ß responses that also activated and protected distal brain areas. Our results are surprising because in other infection models astrocytes have not yet been identified as major type I IFN producers.


Sujet(s)
Astrocytes/immunologie , Encéphalite virale/immunologie , Interféron bêta/métabolisme , Bulbe olfactif/immunologie , Infections à Rhabdoviridae/immunologie , Vesiculovirus/immunologie , Animaux , Astrocytes/virologie , Modèles animaux de maladie humaine , Analyse de profil d'expression de gènes , Gènes rapporteurs , Protéines à fluorescence verte/analyse , Protéines à fluorescence verte/génétique , Interféron bêta/déficit , Luciferases/analyse , Luciferases/génétique , Souris de lignée C57BL , Souris knockout , Neurones/immunologie , Neurones/virologie , Bulbe olfactif/virologie , Analyse de survie
7.
J Interferon Cytokine Res ; 33(11): 660-71, 2013 Nov.
Article de Anglais | MEDLINE | ID: mdl-23848523

RÉSUMÉ

Effective monitoring of the development of neutralizing antibodies (NAbs) against IFN-ß in multiple sclerosis (MS) patients on IFN-ß therapy is important for clinical decision making and disease management. To date, antiviral assays have been the favored approach for NAb determination, but variations in assay conditions between laboratories and the increasing use of novel assays have contributed to the reporting of inconsistent antibody data between laboratories and between products. This study, undertaken at the request of the Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency (EMA), is a joint effort by manufacturers of IFN-ß products (approved in Europe) towards harmonization of a NAb assay that facilitates generation of comparable NAb data, which, in conjunction with clinical outcomes, should prove useful for clinicians treating MS patients with IFN-ß products. This article describes the standardized cellular myxovirus resistance protein A (MxA) protein measurement-based assay for detection of IFN-ß NAbs and its use for the validation of assays used for the quantitative determination of such antibodies. Although titers varied between laboratories and the products used, utilization of IFN-ß1a rather than IFN-ß1b as the challenge antigen produced more consistent results in the NAb assay. Adoption of the standardized assay improves comparability between laboratories circumventing problems that arise when different, nonstandardized assays are employed for immunogenicity assessment. Based on the data, the EMA recommended for standardization purposes, the use of IFN-ß1a in NAb assays, independent of the therapeutic product used for therapy and validation of new NAb procedures against the standardized assay described.


Sujet(s)
Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Interféron bêta/sang , Interféron bêta/immunologie , Sclérose en plaques/sang , Sclérose en plaques/immunologie , Protéines de résistance aux myxovirus/sang , Humains , Interféron bêta/usage thérapeutique , Sclérose en plaques/traitement médicamenteux , Protéines de résistance aux myxovirus/immunologie , Normes de référence
8.
Ther Drug Monit ; 35(4): 530-8, 2013 Aug.
Article de Anglais | MEDLINE | ID: mdl-23765033

RÉSUMÉ

BACKGROUND: Several techniques are used to measure infliximab (IFX) and anti-IFX antibodies (Abs) in Crohn's disease. The aim of this study was to compare different assays for this purpose. METHODS: Fluid-phase radioimmunoassay (RIA), solid-phase enzyme-linked immunosorbent assay (ELISA), reporter gene assay (RGA), and enzyme immunoassay (EIA; anti-IFX Ab only) were assessed. IFX was added to pooled serum from 13 patients with inactive Crohn's disease to yield concentrations of 0, 1, 3, and 9 µg/mL. Anti-IFX Abs were assessed in 6 patients. RESULTS: IFX assessments: RIA and RGA had lower limit of detection than ELISA (0.07 µg/mL and 0.13 versus 0.26). Maximal inaccuracies were 39%, 24%, and 23%. Imprecisions (coefficients of variation) were ≤20% within IFX concentrations between 1 and 9 µg/mL. All assays showed linear correlations (R = 0.97-0.99), but sample concentrations differed by up to 1.55 µg/mL for RIA and RGA, 1.41 µg/mL for ELISA and RIA, and 0.48 µg/mL for ELISA and RGA (P < 0.05). Anti-IFX Ab assessments: RGA gave highly reproducible results (coefficients of variation ≤ 7%) compared with all others (24%-26%). All assays had linear correlations (R = 0.71-0.93), except ELISA versus RGA and EIA. Assays disagreed on anti-IFX Ab titers with mean difference -420 (-1200 to 210) in RGA and EIA, and up to 4500 (-2700 to 11,800) in RIA and RGA. A contributing factor to these discrepancies was inability of ELISA to detect IgG4 anti-IFX Abs. CONCLUSIONS: Performances of assays for IFX and anti-IFX Abs are comparable. However, IFX concentrations and anti-IFX Ab titers show systematic differences, and in individual patients, only the same assay should be used. Problems may arise when different assays are used to manage therapies in the same patient.


Sujet(s)
Anticorps monoclonaux/analyse , Anticorps monoclonaux/immunologie , Anticorps/analyse , Maladie de Crohn/traitement médicamenteux , Maladie de Crohn/immunologie , Test ELISA/méthodes , Dosage radioimmunologique/méthodes , Anticorps/sang , Anticorps/composition chimique , Anticorps monoclonaux/sang , Anticorps monoclonaux/usage thérapeutique , Maladie de Crohn/sang , Surveillance des médicaments/méthodes , Humains , Infliximab , Limite de détection
9.
Bioanalysis ; 4(17): 2179-90, 2012 Sep.
Article de Anglais | MEDLINE | ID: mdl-23013400

RÉSUMÉ

Biopharmaceuticals are used extensively for the treatment of a number of chronic debilitating and fatal diseases such as cancer and inflammatory or autoimmune diseases. Although biopharmaceuticals are in general well tolerated, the development of anti-drug antibodies can impair their safety and efficacy. Assessment of immunogenicity is essential for a more effective and rational use of biopharmaceuticals, and is dependent upon the establishment of efficient standardized assays that allow direct comparison of immunogenicity data with clinical outcome. Although regulatory authorities recommend the use of cell-based assays that reflect the mechanism of action of the drug for the detection of neutralizing anti-drug antibodies, conventional cell-based assays are difficult to standardize and often give variable results. A number of strategies have been adopted to improve the performance of cell-based assays, including quantification of drug-induced proteins using either real-time RT-PCR or branched DNA to detect mRNA, or ELISAs to detect protein, bridging assays using immobilized cells and the use of reporter gene assays. The relative merits and limitations of each of these methods is reviewed herein.


Sujet(s)
Anticorps neutralisants/analyse , Biopharmacie/normes , Anticorps neutralisants/immunologie , Biopharmacie/tendances , Lignée cellulaire tumorale , Humains
10.
Proc Natl Acad Sci U S A ; 109(26): 10486-91, 2012 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-22689946

RÉSUMÉ

The synthetic double-stranded RNA poly(I:C) is commonly used as an adjuvant to boost CD8 T-cell function; however, polyinosinic:polycytidylic acid [poly(I:C)] can also suppress autoimmune disease. The mechanism by which a single adjuvant achieves two distinct immunoregulatory roles is unknown. Although it is clear that coadministration of poly(I:C) with antigen elicits strong adjuvant effects in mice, we found that poly(I:C) injection before antigen substantially reduced antigen-dependent CD8 T-cell responses. Notably, CD8 T cells sensitized in poly(I:C)-pretreated mice failed to fully up-regulate IL-33R (ST2), which led to impaired T-cell receptor-independent responses to IL-33. In contrast, nonsensitized effector CD8 T cells responded robustly to IL-33 using a two-signal cytokine mechanism. During an acute lung response to Staphylococcus aureus enterotoxin, peripheral injection of poly(I:C) manifested a suppressive process by inhibiting the differentiation of both antigen- and IL-33-responsive CD8 effectors systemically. These findings highlight that early exposure to double-stranded RNA reverses its role as an adjuvant and, importantly, prevents IL-33R up-regulation on CD8 effector T cells to dampen inflammation.


Sujet(s)
Lymphocytes T CD8+/cytologie , Différenciation cellulaire/physiologie , Interleukines/physiologie , Récepteur de type Toll-3/métabolisme , Animaux , Antigènes/immunologie , Lymphocytes T CD8+/immunologie , Interleukine-33 , Ligands , Activation des lymphocytes , Souris , ARN double brin/administration et posologie
11.
Expert Rev Clin Pharmacol ; 4(5): 623-31, 2011 Sep.
Article de Anglais | MEDLINE | ID: mdl-22114889

RÉSUMÉ

The safety and efficacy of biopharmaceuticals can be severely impaired by their immunogenicity. A risk-based strategy should be used to assess immunogenicity on a case-by-case basis using standardized methods to correlate anti-drug antibody levels with clinical outcome. In silico and in vitro techniques allow putative T-cell epitopes to be identified and eliminated in candidate molecules while maintaining structure and function. Putative T-cell epitopes can be studied in the context of the HLA allotypes representative of the target population in vitro and in transgenic mice that express human HLA genes. Mice immune tolerant to human proteins allow the study of the effect of factors such as aggregation on the loss of immune tolerance. However, significant challenges remain in order to be able predict the immunogenicity of a therapeutic protein in a particular individual.


Sujet(s)
Produits biologiques/immunologie , Produits biologiques/pharmacologie , Phénomènes immunogénétiques/effets des médicaments et des substances chimiques , Phénomènes immunogénétiques/immunologie , Animaux , Déterminants antigéniques des lymphocytes T/effets indésirables , Déterminants antigéniques des lymphocytes T/immunologie , Humains , Tolérance immunitaire
12.
J Immunol Methods ; 373(1-2): 229-39, 2011 Oct 28.
Article de Anglais | MEDLINE | ID: mdl-21910993

RÉSUMÉ

A cell-based assay has been developed for the quantification of the activity of TNFα antagonists based on human erythroleukemic K562 cells transfected with a NFκB regulated firefly luciferase reporter-gene construct. Both drug activity and anti-drug neutralizing antibodies can be quantified with a high degree of precision within 2h, and without interference from cytokines and other factors known to activate NFκB. The assay cells also contain the Renilla luciferase reporter gene under the control of a constitutive promoter that allows TNFα-induced firefly luciferase activity to be normalized relative to Renilla luciferase expression. Thus, results are independent of cell number or differences in cell viability, resulting in intra and inter assay coefficients of variation of 10% or less. Normalization of results relative to the expression of an internal standard also provides a means for correcting for serum matrix effects and allows residual drug levels or anti-drug neutralizing antibodies to be quantified even in serum samples with a relatively high degree of cytotoxicity.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Anticorps neutralisants/analyse , Luciferases/métabolisme , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs , Adalimumab , Animaux , Anticorps monoclonaux/sang , Anticorps monoclonaux/immunologie , Anticorps monoclonaux humanisés/sang , Anticorps monoclonaux humanisés/immunologie , Anticorps monoclonaux humanisés/pharmacologie , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Polyarthrite rhumatoïde/sang , Polyarthrite rhumatoïde/traitement médicamenteux , Maladie de Crohn/sang , Maladie de Crohn/traitement médicamenteux , Milieux de culture/composition chimique , Milieux de culture/pharmacologie , Relation dose-effet des médicaments , Régulation de l'expression des gènes codant pour des enzymes/effets des médicaments et des substances chimiques , Humains , Infliximab , Cellules K562 , Luciferases/génétique , Luciférases des lucioles/génétique , Luciférases des lucioles/métabolisme , Luciférases de Renilla/génétique , Luciférases de Renilla/métabolisme , Facteur de transcription NF-kappa B/génétique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Séquences d'acides nucléiques régulatrices/génétique , Sérum/composition chimique , Sérum/immunologie , Facteur de nécrose tumorale alpha/immunologie , Facteur de nécrose tumorale alpha/pharmacologie
13.
Ther Adv Drug Saf ; 2(3): 113-28, 2011 Jun.
Article de Anglais | MEDLINE | ID: mdl-25083207

RÉSUMÉ

Biopharmaceuticals are used widely for the treatment of cancer, chronic viral hepatitis, inflammatory, and autoimmune diseases. Biopharmaceuticals such as interferons are well tolerated for the most part with the most common adverse events observed being 'flu-like' symptoms that resolve rapidly after initial treatment. Prolonged treatment is associated, however, with more serious adverse events including leucopenia, thrombocytopenia, and neuropsychiatric effects, which may necessitate dose reduction or even cessation of treatment in some patients. Recombinant growth factors, such as erythropoietin (EPO), granulocyte colony-stimulating factor, or granulocyte macrophage colony-stimulating factor, are for the most part well tolerated, although severe complications have been reported in patients with cancer or chronic kidney disease treated with EPO. Similarly, treatment of patients with cancer with high doses of interleukin-2 is associated with significant toxicity. Treatment of chronic inflammatory diseases, such as rheumatoid arthritis, psoriasis, and Crohn's disease, with antitumor necrosis factor-alpha monoclonal antibodies is associated with an increased risk of granulomatous infections and, in particular, tuberculosis. The monoclonal antibody, natalizumab, that targets alpha4 integrins is effective in the treatment of multiple sclerosis but is associated with the activation of JC virus and development of progressive multifocal leukoencephalopathy. Repeated administration of recombinant proteins can cause a break in immune tolerance in some patients resulting in the production of a polyclonal antibody response that can adversely affect pharmacokinetics and clinical response. In addition, neutralizing antibodies that cross react with nonredundant essential proteins such as EPO can cause severe autoimmune reactions.

18.
Eur J Immunol ; 40(10): 2769-77, 2010 Oct.
Article de Anglais | MEDLINE | ID: mdl-20821729

RÉSUMÉ

Virus-induced expansion of CD8(+) T cells may be promoted by type I IFN receptor (IFNAR)-triggering of T cells, depending on the pathogen tested. We studied modified vaccinia virus Ankara (MVA), a promising vaccine vector candidate, which was derived from conventional vaccinia virus (VACV) by more than 570 consecutive in vitro passages. In adoptive transfer experiments, we verified that VACV expressing the gp33 epitope of lymphocytic choriomeningitis virus (VACV(gp33)) induced largely IFNAR-independent expansion of gp33-specific T cells. On the contrary, MVA(gp33)-induced T-cell expansion was IFNAR dependent. Interestingly, under the latter conditions, T-cell activation was IFNAR independent, whereas T-cell apoptosis was enhanced in the absence of IFNAR. To address whether MVA-induced T-cell expansion was solely affected by IFNAR-triggering of T cells, expansion of endogenous T cells was studied in conditional mice with a T-cell- or DC-specific IFNAR deletion. Interestingly, both mouse strains showed moderately reduced T-cell expansion, whereas mice with a combined T-cell- and DC-specific IFNAR ablation showed massively reduced T-cell expansion similar to that of IFNAR(-/-) mice. These results are compatible with the model that IFN-inducing viruses such as MVA confer virus-specific CD8(+) T-cell expansion by concomitant IFNAR-triggering of DC and of T cells.


Sujet(s)
Lymphocytes T CD8+/immunologie , Cellules dendritiques/immunologie , Vecteurs génétiques/immunologie , Interféron de type I/immunologie , Virus de la vaccine/immunologie , Transfert adoptif , Animaux , Antigènes CD11c/immunologie , Déterminants antigéniques des lymphocytes T , Cytométrie en flux , Activation des lymphocytes/immunologie , Souris , Transduction du signal/immunologie
19.
J Virol ; 84(13): 6549-63, 2010 Jul.
Article de Anglais | MEDLINE | ID: mdl-20410285

RÉSUMÉ

Type I interferons (IFNs) are considered to be important mediators of innate immunity due to their inherent antiviral activity, ability to drive the transcription of a number of genes involved in viral clearance, and their role in the initiation of innate and adaptive immune responses. Due to the central role of type I IFNs, we sought to determine their importance in the generation of immunity to a recombinant vaccine vector fowlpox virus (FPV). In analyzing the role of type I IFNs in immunity to FPV, we show that they are critical to the secretion of a number of innate and proinflammatory cytokines, including type I IFNs themselves as well as interleukin-12 (IL-12), tumor necrosis factor-alpha (TNF-alpha), IL-6, and IL-1beta, and that deficiency leads to enhanced virus-mediated antigen expression. Interestingly, however, type I IFNs were not required for adaptive immune responses to recombinant FPV even though plasmacytoid dendritic cells (pDCs), the primary producers of type I IFNs, have been shown to be requisite for this to occur. Furthermore, we provide evidence that the importance of pDCs may lie in their ability to capture and present virally derived antigen to T cells rather than in their capacity as professional type I IFN-producing cells.


Sujet(s)
Immunité acquise , Cytokines/immunologie , Cellules dendritiques/immunologie , Virus de la variole de la volaille/immunologie , Interféron de type I/immunologie , Animaux , Cellules présentatrices d'antigène/immunologie , Antigènes viraux/immunologie , Souris , Souris de lignée C57BL , Lymphocytes T/immunologie , Vaccins synthétiques/immunologie , Vaccins antiviraux/immunologie
20.
J Immunol Methods ; 356(1-2): 18-28, 2010 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-20298696

RÉSUMÉ

Assessment of immunogenicity is an important part of biopharmaceutical drug safety evaluation and a prerequisite for the development of less immunogenic and safer biopharmaceuticals since anti-drug antibodies can impair the activity and compromise the safety of biopharmaceuticals. Although regulatory authorities recommend cell-based assays for detection of neutralizing antibodies (NAbs), such assays are difficult to standardize, and ill adapted to high-throughput analysis. These limitations have been overcome by the development of a unique one-step cell-based assay that allows both drug activity and drug NAbs to be quantified rapidly and with a high degree of precision simply be adding reporter cells to a sample. The reporter cells have been engineered to express firefly luciferase (FL) under the control of a drug-responsive promoter, and to express the drug of interest, the production of which is normalized relative to the expression of Renilla luciferase (RL) transcribed from a common doxycycline-inducible promoter. Residual drug levels present in a sample are first quantified by determination of FL expression, autocrine drug synthesis is then induced, and NAb activity is quantified from the difference in the ratio of FL/RL expression in the presence or absence of the sample. Since assay results are normalized relative to the expression of an internal standard, results are independent of cell number or differences in cell viability thus affording a high degree of assay precision and reducing serum matrix effects to a minimum. This unique assay platform is ideally suited for high-throughput analysis, is applicable to most biopharmaceuticals, and will facilitate standardization and comparison of immunogenicity data. The performance of the one-step assay is illustrated for interferon alpha2 (IFNalpha2) used widely to treated chronic hepatitis C (HCV) infection and neoplastic disease.


Sujet(s)
Anticorps neutralisants/analyse , Techniques génétiques , Mesures de luminescence/méthodes , Anticorps neutralisants/immunologie , Lignée cellulaire tumorale , Gènes rapporteurs , Humains , Interférons/sang , Interférons/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE