982 resultados para humanized mice, bispecific antibodies, MCSP


Relevância:

100.00% 100.00%

Publicador:

Resumo:

Humanes MCSP ist ein gut charakterisiertes Tumorantigen, das auf der Mehrzahl aller malignen Melanome hoch exprimiert wird, und stellt somit eine gute Zielstruktur für immuntherapeutische Ansätze dar. rnInnerhalb der vorliegenden Arbeit wurden die Wirkmechanismen eines neuen bispezifischen Antikörpers, der gegen humanes MCSP und CD3 auf T-Zellen gerichtet ist, in vitro und im humanisierten Tumormausmodell in vivo untersucht. In humanen T Zellkokulturen induzierte der bispezifische MCSP-CD3 Antikörper in Gegenwart MCSP-positiver Melanomzellen konzentrationsabhängige T-Zellaktivierung, Sekretion von Zytokinen und effiziente Tumorzelllyse durch CD4- und CD8-positive T-Zellen. Die induzierte Lyse war hierbei unabhängig von der T-Zellrezeptorspezifität sowie kostimulatorischen Molekülen und allein abhängig von der Expression des Tumorantigens sowie CD3 auf den T-Zellen. Wie hier diskutiert, liegt es nahe, dass die Freisetzung lytischer Moleküle (Perforin und Granzym-B) durch CD8- und auch CD4 positiver T-Zellen den Hauptmechanismus in der Lyse der Melanomzellen darstellt. rnUm die Wirksamkeit in vivo testen zu können, wurde ein humanisiertes Tumormausmodell etabliert. Die Injektion humaner hämatopoetischer Stammzellen in neugeborene Rag2-/-gc-/- Mäuse führte zur Entwicklung funktioneller T-Zellen im murinen Thymus, welche lymphatische Organe besiedelten. In vitro induzierten die T-Zellen humanisierter Mäuse in Anwesenheit des bispezifischen MCSP-CD3 Antikörpers ebenfalls konzentrationsabhängige Lyse der Melanomzellen. Wie hier gezeigt, induzierte die Injektion humaner Melanomzellen in humanisierte Mäuse keine messbare Abstoβungsreaktion. Unter Behandlung mit MCSP-CD3 wurde zwar eine erhöhte Anzahl humaner T-Zellen im Tumorgewebe nachgewiesen, jedoch verfügte die verwendete Melanomzelllinie über eine geringe basale T Zellinfiltration, geringe Vaskularisierung und ein noduläres Wachstumsverhalten. Wie innerhalb dieser Arbeit diskutiert, kann durch die Kombination mit Therapien, die eine erhöhte T-Zellinfiltration in das Tumorgewebe ermöglichen, die Wirksamkeit von bispezifischen Antikörpern möglicherweise gesteigert werden. rn

Relevância:

100.00% 100.00%

Publicador:

Resumo:

The pregnane X receptor (PXR) has been postulated to play a role in the metabolism of α-tocopherol owing to the up-regulation of hepatic cytochrome P450 (P450) 3A in human cell lines and murine models after α-tocopherol treatment. However, in vivo studies confirming the role of PXR in α-tocopherol metabolism in humans presents significant difficulties and has not been performed. PXR-humanized (hPXR), wild-type, and Pxr-null mouse models were used to determine whether α-tocopherol metabolism is influenced by species-specific differences in PXR function in vivo. No significant difference in the concentration of the major α-tocopherol metabolites was observed among the hPXR, wild-type, and Pxr-null mice through mass spectrometry-based metabolomics. Gene expression analysis revealed significantly increased expression of Cyp3a11 as well as several other P450s only in wild-type mice, suggesting species-specificity for α-tocopherol activation of PXR. Luciferase reporter assay confirmed activation of mouse PXR by α-tocopherol. Analysis of the Cyp2c family of genes revealed increased expression of Cyp2c29, Cyp2c37, and Cyp2c55 in wild-type, hPXR, and Pxr-null mice, which suggests PXR-independent induction of Cyp2c gene expression. This study revealed that α-tocopherol is a partial agonist of PXR and that PXR is necessary for Cyp3a induction by α-tocopherol. The implications of a novel role for α-tocopherol in Cyp2c gene regulation are also discussed.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Due to multiple immune evasion mechanisms of cancer cells, novel therapy approaches are required to overcome the limitations of existing immunotherapies. Bispecific antibodies are potent anti-cancer drugs, which redirect effector T cells for specific tumor cell lysis, thus enabling the patient’s immune system to fight cancer cells. The antibody format used in this proof of concept study–bispecific ideal monoclonal antibodies termed BiMAB–is a tailor-made recombinant protein, which consists of two fused scFv antibodies recognizing different antigens. Both are arranged in tandem on a single peptide chain and the individual variable binding domains are separated by special non-immunogenic linkers. The format is comprised of a scFv targeting CLDN18.2–a gastric cancer tumor associated antigen (TAA) –while the second specificity binds the CD3 epsilon (CD3ε) subunit of the T cell receptor (TCR) on T cells. For the first time, we compared in our IMAB362-based BiMAB setting, four different anti-CD3-scFvs, respectively derived from the mAbs TR66, CLB-T3, as well as the humanized and the murine variant of UCHT1. In addition, we investigated the impact of an N- versus a C-terminal location of the IMAB362-derived scFv and the anti-CD3-scFvs. Thus, nine CLDN18.2 specific BiMAB proteins were generated, of which all showed a remarkably high cytotoxicity towards CLDN18.2-positive tumor cells. Because of its promising effectiveness, 1BiMAB emerged as the BiMAB prototype. The selectivity of 1BiMAB for its TAA and CD3ε, with affinities in the nanomolar range, has been confirmed by in vitro assays. Its dual binding depends on the design of an N-terminally positioned IMAB362 scFv and the consecutive C-terminally positioned TR66 scFv. 1BiMAB provoked a concentration and target cell dependent T cell activation, proliferation, and upregulation of the cytolytic protein Granzyme B, as well as the consequent elimination of target cells. Our results demonstrate that 1BiMAB is able to activate T cells independent of elements that are usually involved in the T cell recognition program, like antigen presentation, MHC restriction, and co-stimulatory effector molecules. In the first in vivo studies using a subcutaneous xenogeneic tumor mouse model in immune incompetent NSG mice, we could prove a significant therapeutic effect of 1BiMAB with partial or complete tumor elimination. The initial in vitro RIBOMAB experiments correspondingly showed encouraging results. The electroporation of 1BiMAB IVT-RNA into target or effector cells was feasible, while the functionality of translated 1BiMAB was proven by induced T cell activation and target cell lysis. Accordingly, we could show that the in vitro RIBOMAB approach was applicable for all nine BiMABs, which proves the RIBOMAB concept. Thus, the CLDN18.2-BiMAB strategy offers great potential for the treatment of cancer. In the future, administered either as protein or as IVT-RNA, the BiMAB format will contribute towards finding solutions to raise and sustain tumor-specific cellular responses elicited by engaged and activated endogenous T cells. This will potentially enable us to overcome immune evasion mechanisms of tumor cells, consequently supporting current solid gastric cancer therapies.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Despite positive testing in animal studies, more than 80% of novel drug candidates fail to proof their efficacy when tested in humans. This is primarily due to the use of preclinical models that are not able to recapitulate the physiological or pathological processes in humans. Hence, one of the key challenges in the field of translational medicine is to “make the model organism mouse more human.” To get answers to questions that would be prognostic of outcomes in human medicine, the mouse's genome can be altered in order to create a more permissive host that allows the engraftment of human cell systems. It has been shown in the past that these strategies can improve our understanding of tumor immunology. However, the translational benefits of these platforms have still to be proven. In the 21st century, several research groups and consortia around the world take up the challenge to improve our understanding of how to humanize the animal's genetic code, its cells and, based on tissue engineering principles, its extracellular microenvironment, its tissues, or entire organs with the ultimate goal to foster the translation of new therapeutic strategies from bench to bedside. This article provides an overview of the state of the art of humanized models of tumor immunology and highlights future developments in the field such as the application of tissue engineering and regenerative medicine strategies to further enhance humanized murine model systems.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Animal models have been relevant to study the molecular mechanisms of cancer and to develop new antitumor agents. Anyway, the huge divergence in mouse and human evolution made difficult the translation of the gained achievements in preclinical mouse based studies. The generation of clinically relevant murine models requires their humanization both concerning the creation of transgenic models and the generation of humanized mice in which to engraft a functional human immune system, and reproduce the physiological effects and molecular mechanisms of growth and metastasization of human tumors. In particular, the availability of genotypically stable immunodepressed mice able to accept tumor injection and allow human tumor growth and metastasization would be important to develop anti-tumor and anti-metastatic strategies. Recently, Rag2-/-;gammac-/- mice, double knockout for genes involved in lymphocyte differentiation, had been developed (CIEA, Central Institute for Experimental Animals, Kawasaki, Japan). Studies of human sarcoma metastasization in Rag2-/-; gammac-/- mice (lacking B, T and NK functionality) revealed their high metastatic efficiency and allowed the expression of human metastatic phenotypes not detectable in the conventionally used nude murine model. In vitro analysis to investigate the molecular mechanisms involved in the specific pattern of human sarcomas metastasization revealed the importance of liver-produced growth and motility factors, in particular the insulin-like growth factors (IGFs). The involvement of this growth factor was then demonstrated in vivo through inhibition of IGF signalling pathway. Due to the high growth and metastatic propensity of tumor cells, Rag2-/-;gammac-/- mice were used as model to investigate the metastatic behavior of rhabdomyosarcoma cells engineered to improve the differentiation. It has been recently shown that this immunodeficient model can be reconstituted with a human immune system through the injection of human cord blood progenitor cells. The work illustrated in this thesis revealed that the injection of different human progenitor cells (CD34+ or CD133+) showed peculiar engraftment and differentiation abilities. Experiments of cell vaccination were performed to investigate the functionality of the engrafted human immune system and the induction of specific human immune responses. Results from such experiments will allow to collect informations about human immune responses activated during cell vaccination and to define the best reconstitution and experimental conditions to create a humanized model in which to study, in a preclinical setting, immunological antitumor strategies.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

The obligate intracellular pathogen Chlamydia trachomatis is a gram negative bacterium which infects epithelial cells of the reproductive tract. C. trachomatis is the leading cause of bacterial sexually transmitted disease worldwide and a vaccine against this pathogen is highly needed. Many evidences suggest that both antigen specific-Th1 cells and antibodies may be important to provide protection against Chlamydia infection. In a previous study we have identified eight new Chlamydia antigens inducing CD4-Th1 and/or antibody responses that, when combined properly, can protect mice from Chlamydia infection. However, all selected recombinant antigens, upon immunization in mice, elicited antibodies not able to neutralize Chlamydia infectivity in vitro. With the aim to improve the quality of the immune response by inducing effective neutralizing antibodies, we used a novel delivery system based on the unique capacity of E. coli Outer Membrane Vesicles (OMV) to present membrane proteins in their natural composition and conformation. We have expressed Chlamydia antigens, previously identified as vaccine candidates, in the OMV system. Among all OMV preparations, the one expressing HtrA Chlamydia antigen (OMV-HtrA), showed to be the best in terms of yield and quantity of expressed protein, was used to produce mice immune sera to be tested in neutralization assay in vitro. We observed that OMV-HtrA elicited specific antibodies able to neutralize efficiently Chlamydia infection in vitro, indicating that the presentation of the antigens in their natural conformation is crucial to induce an effective immune response. This is one of the first examples in which antibodies directed against a new Chlamydia antigen, other than MOMP (the only so far known antigen inducing neutralizing antibodies), are able to block the Chlamydia infectivity in vitro. Finally, by performing an epitope mapping study, we investigated the specificity of the antibody response induced by the recombinant HtrA and by OMV-HtrA. In particular, we identified some linear epitopes exclusively recognized by antibodies raised with the OMV-HtrA system, detecting in this manner the antigen regions likely responsible of the neutralizing effect.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Die Multiple Sklerose (MS) ist eine Autoimmunkrankheit des zentralen Nervensystems, bei der sich autoreaktive T-Effektorzellen der Kontrolle durch regulatorische T-Zellen (Treg) entziehen. Innerhalb dieser Arbeit wurde gezeigt, dass T-Effektorzellen von MS-Patienten insensitiv gegenüber der Suppression durch Treg sind. Hervorgerufen wird diese Treg-Resistenz durch Interleukin-6 (IL-6). Die Inhibition des IL-6-Signalweges stellt die Treg-vermittelte Suppression der T-Effektorzellen wieder her. Es zeigte sich, dass die Bildung von IL-6 und die Expression des IL-6-Rezeptors in MS-Patienten in einer positiven Rückkopplungsschleife von IL-6 selbst induziert werden.rnZur Analyse humaner Immunantworten in vivo und deren Modulation durch humanspezifische Therapeutika wurden humanisierte Mausmodelle etabliert. Der adoptive Transfer humaner Immunzellen in immundefiziente Mäuse erlaubte die Untersuchung von T-Lymphozyten, die aus dem Blut von MS-Patienten isoliert wurden. Es zeigte sich, dass Treg-resistente T-Effektorzellen aus den MS-Patienten in den Tieren eine letale Graft-versus-Host-Erkrankung auslösten, die nicht durch aktivierte Treg therapiert werden konnte. Erst eine Behandlung mit dem humanspezifischen anti-IL-6-Antikörper Tocilizumab in vivo konnte die Erkrankung der Tiere deutlich abmildern.rnIm zweiten Modell wurden immundefiziente Mäuse mit humanen CD34+ Blutstammzellen immunologisch rekonstituiert. Diese Tiere entwickelten ein nahezu vollständig humanes Immunsystem. Die Immunisierung mit dem murinen Myelin-Oligodenrozyten-Glykoprotein löste in den humanisierten Mäusen eine MS-ähnliche Autoimmunität aus. Die Neuroinflammation wurde durch humane T- und B-Zellen vermittelt, korrelierte mit erhöhter IL-17-Produktion und führte zu einer IL-6-abhängigen Treg-Resistenz der T-Effektorzellen. Somit eignen sich die etablierten Modelle, um zukünftig die Wirksamkeit neuer Therapeutika zur Behandlung der MS präklinisch zu testen.rn

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Procainamide, a type I antiarrhythmic agent, is used to treat a variety of atrial and ventricular dysrhythmias. It was reported that long-term therapy with procainamide may cause lupus erythematosus in 25-30% of patients. Interestingly, procainamide does not induce lupus erythematosus in mouse models. To explore the differences in this side-effect of procainamide between humans and mouse models, metabolomic analysis using ultra-performance liquid chromatography coupled with electrospray ionization quadrupole time-of-flight mass spectrometry (UPLC-ESI-QTOFMS) was conducted on urine samples from procainamide-treated humans, CYP2D6-humanized mice, and wild-type mice. Thirteen urinary procainamide metabolites, including nine novel metabolites, derived from P450-dependent, FMO-dependent oxidations and acylation reactions, were identified and structurally elucidated. In vivo metabolism of procainamide in CYP2D6-humanized mice as well as in vitro incubations with microsomes and recombinant P450s suggested that human CYP2D6 plays a major role in procainamide metabolism. Significant differences in N-acylation and N-oxidation of the drug between humans and mice largely account for the interspecies differences in procainamide metabolism. Significant levels of the novel N-oxide metabolites produced by FMO1 and FMO3 in humans might be associated with the development of procainamide-induced systemic lupus erythematosus. Observations based on this metabolomic study offer clues to understanding procainamide-induced lupus in humans and the effect of P450s and FMOs on procainamide N-oxidation.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Approximately one-third of the world's population suffers from chronic helminth infections with no effective vaccines currently available. Antibodies and alternatively activated macrophages (AAM) form crucial components of protective immunity against challenge infections with intestinal helminths. However, the mechanisms by which antibodies target these large multi-cellular parasites remain obscure. Alternative activation of macrophages during helminth infection has been linked to signaling through the IL-4 receptor alpha chain (IL-4Rα), but the potential effects of antibodies on macrophage differentiation have not been explored. We demonstrate that helminth-specific antibodies induce the rapid trapping of tissue migrating helminth larvae and prevent tissue necrosis following challenge infection with the natural murine parasite Heligmosomoides polygyrus bakeri (Hp). Mice lacking antibodies (JH (-/-)) or activating Fc receptors (FcRγ(-/-)) harbored highly motile larvae, developed extensive tissue damage and accumulated less Arginase-1 expressing macrophages around the larvae. Moreover, Hp-specific antibodies induced FcRγ- and complement-dependent adherence of macrophages to larvae in vitro, resulting in complete larval immobilization. Antibodies together with helminth larvae reprogrammed macrophages to express wound-healing associated genes, including Arginase-1, and the Arginase-1 product L-ornithine directly impaired larval motility. Antibody-induced expression of Arginase-1 in vitro and in vivo occurred independently of IL-4Rα signaling. In summary, we present a novel IL-4Rα-independent mechanism of alternative macrophage activation that is antibody-dependent and which both mediates anti-helminth immunity and prevents tissue disruption caused by migrating larvae.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

High-affinity folate receptors (FRs) are expressed at elevated levels on many human tumors. Bispecific antibodies that bind the FR and the T-cell receptor (TCR) mediate lysis of these tumor cells by cytotoxic T lymphocytes. In this report, conjugates that consist of folate covalently linked to anti-TCR antibodies are shown to be potent in mediating lysis of tumor cells that express either the alpha or beta isoform of the FR. Intact antibodies with an average of five folate per molecule exhibited high affinity for FR+ tumor cells but did not bind to FR- tumor cells. Lysis of FR+ cell lines could be detected at concentrations as low as 1 pM (approximately 0.1 ng/ml), which was 1/1000th the concentration required to detect binding to the FR+ cells. Various FR+ mouse tumor cell lines could be targeted with each of three different anti-TCR antibodies that were tested as conjugates. The antibodies included 1B2, a clonotypic antibody specific for the cytotoxic T cell clone 2C; KJ16, an anti-V beta 8 antibody; and 2C11, an anti-CD3 antibody. These antibodies differ in affinities by up to 100-fold, yet the cytolytic capabilities of the folate/antibody conjugates differed by no more than 10-fold. The reduced size (in comparison with bispecific antibodies) and high affinity of folate conjugates suggest that they may be useful as immunotherapeutic agents in targeting tumors that express folate receptors.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Construction of a bispecific single-chain antibody derivative is described that consists of two different single-chain Fv fragments joined through a Gly-Ser linker. One specificity of the two Fv fragments is directed against the CD3 antigen of human T cells and the other is directed against the epithelial 17-1A antigen; the latter had been found in a clinical trial to be a suitable target for antibody therapy of minimal residual colorectal cancer. The construct could be expressed in CHO cells as a fully functional protein, while its periplasmic expression in Escherichia coli resulted in a nonfunctional protein only. The antigen-binding properties of the bispecific single-chain antibody are indistinguishable from those of the corresponding univalent single-chain Fv fragments. By redirecting human peripheral T lymphocytes against 17-1A-positive tumor cells, the bispecific antibody proved to be highly cytotoxic at nanomolar concentrations as demonstrated by 51Cr release assay on various cell lines. The described bispecific construct has a molecular mass of 60 kDa and can be easily purified by its C-terminal histidine tail on a Ni-NTA chromatography column. As bispecific antibodies have already been shown to be effective in vivo in experimental tumor systems as well as in phase-one clinical trials, the small CD3/17-1A-bispecific antibody may be more efficacious than intact antibodies against minimal residual cancer cells.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Three distinct isolates of Candida albicans were used to establish systemic and oral infections in inbred mice that are genetically resistant or susceptible to tissue damage. Patterns of infection differed significantly between both yeasts and mouse strains. Systemic infection conferred significant protection against re-challenge with the homologous, but not the heterologous yeast; however, the protective effect was more evident in the tissue-susceptible CBA/CaH mice than in the resistant BALB/c strain. In contrast, oral infection induced protection against both homologous and heterologous oral challenge, although this was significant only in the CBA/CaH mice. CBA/CaH mice produced antibodies of both IgG1 and IgG2a subclasses, whereas BALB/c mice produced predominantly IgG1. Western blotting demonstrated considerable differences between epitopes recognised by serum antibodies from mice of both strains after immunisation with each of the three yeasts. Thus, different strains of yeast show considerable specificity in antibody responses elicited by either systemic or oral infection. (c) 2005 Elsevier SAS. All rights reserved.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

Sporothrix schenckii é um fungo dimórfico e agente etiológico da esporotricose, uma micose profunda que apresenta diferentes manifestações clínicas. As diversas manifestações clínicas desta e de outras doenças infecciosas podem ser relacionadas ao status imune do hospedeiro, a fatores de virulência do patógeno ou a diferentes genótipos. Dados anteriores do nosso grupo demonstram que diferenças na expressão de adesinas do S. schenckii para fibronectina estão diretamente relacionadas à virulência de diferentes cepas. Neste trabalho visamos avaliar caracteres morfológicos bioquímicos e genotípicos de doze isolados de geofílicos, zoofílicos e antropofílicos de S. schenckii, de diferentes origens geográficas, que apresentam diferentes graus de virulência. Foi analisada a morfologia das formas de micélio e levedura de cada isolado. Foi observado que a fase de micélio dos isolados estudados apresentaram morfologia típica com hifas finas e septadas, com conídios obovóides ou ovóides alongados. As leveduras apresentaram pleomorfismo típico da espécie, com células variando do formato ovóide ao alongado. Verificamos ainda a expressão de adesinas para fibronectina e laminina, do antígeno gp70 e, o padrão de bandas antigênicas reconhecidas por anticorpos IgG presentes em soro de pacientes com esporotricose ou de camundongos infectados. Para isso, foram extraídas proteínas de superfície da forma de levedura de cada isolada, sendo os extratos ensaiados por Western blot. Nestes ensaios observamos que os isolados mais virulentos de S. schenckii expressavam mais adesinas para fibronectina e laminina. A presença da gp70 foi detectada em dez dos doze isolados, sendo que apenas os isolados zoofílicos não expressam esta glicoproteína. O padrão antigênico foi variável entre os isolados, não havendo clara relação com a origem e/ou distribuição geográfica. Os dados fenotípicos foram confrontados com dados genotípicos. Para isso, sequenciamos os loci da calmodulina (CAL) e do Internal Transcribed Spacer 1/2 (ITS 1/2) a fim de averiguar se haviam diferenças genotípicas entre os isolados estudados. As análises do sequenciamento do loci CAL e ITS, contudo, apontam a divisão dos isolados em duas espécies filogenéticas, S. schenckii e S. brasiliensis não correlacionada com a distribuição geográfica dos mesmos. Nosso estudo reforça a hipótese de haver uma correlação entre virulência e expressão de adesinas, porém, sem qualquer relação entre a distribuição geográfica dos isolados zoofilicos, antropofílicos ou geofílicos, bem como dos genótipos encontrados.

Relevância:

100.00% 100.00%

Publicador:

Resumo:

La maladie du greffon contre l’hôte (GvHD) est une complication majeure des greffes de cellules souches hématopoïétiques (HSCT) qui survient dans 30 à 70% des cas et peut causer la mort, malgré un traitement prophylactique bien conduit. Il existe donc une réelle demande clinique pour améliorer ces traitements prophylactiques. Parce que ces traitements prophylactiques reposent en général sur des agents immunosuppresseurs, ceux-ci contribuent à diminuer la reconstitution immunitaire du patient, ce qui a un impact défavorable sur les infections et les taux de rechute d’hémopathie maligne, et donc limite leur utilisation. Les immunoglobulines (IVIG) pourraient représenter une alternative intéressante puisqu’elles ont des propriétés immunomodulatrices et qu’elles sont de plus couramment utilisées en clinique pour traiter des patients ayant un déficit immunitaire. Leur capacité à réduire l’apparition et la sévérité de la GvHD, sans toutefois inhiber ou nuire à la reconstitution immunitaire chez le patient n’a néanmoins jamais été clairement démontrée. Les objectifs de ce projet sont donc d’évaluer l’efficacité des IVIG à réduire l’incidence et la sévérité de la GvHD dans un modèle murin humanisé de GvHD, ainsi que de déterminer le mécanisme d’action des IVIG. Ce modèle consiste à injecter des huPBMCs à des souris immunodéprimées ne pouvant les rejeter. Les résultats obtenus suggèrent que les IVIG possèdent un effet immunomodulateur permettant de réduire les signes cliniques et de retarder l’apparition de la GvHD, tout en permettant l’apparition de cellules NK. Les IVIG agiraient de façon indirecte sur les huPBMCs afin d’induire l’apparition des cellules NK.