Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Eur J Immunol ; 51(8): 2074-2085, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33945643

RESUMO

The aberrant release of inflammatory mediators often referred to as a cytokine storm or cytokine release syndrome (CRS), is a common and sometimes fatal complication in acute infectious diseases including Ebola, dengue, COVID-19, and influenza. Fatal CRS occurrences have also plagued the development of highly promising cancer therapies based on T-cell engagers and chimeric antigen receptor (CAR) T cells. CRS is intimately linked with dysregulated and excessive cytokine release, including IFN-γ, TNF-α, IL 1, IL-6, and IL-10, resulting in a systemic inflammatory response leading to multiple organ failure. Here, we show that mice intravenously administered the agonistic hamster anti-mouse CD3ε monoclonal antibody 145-2C11 develop clinical and laboratory manifestations seen in patients afflicted with CRS, including body weight loss, hepatosplenomegaly, thrombocytopenia, increased vascular permeability, lung inflammation, and hypercytokinemia. Blood cytokine levels and gene expression analysis from lung, liver, and spleen demonstrated a hierarchy of inflammatory cytokine production and infiltrating immune cells with differentiating organ-dependent kinetics. IL-2, IFN-γ, TNF-α, and IL-6 up-regulation preceded clinical signs of CRS. The co-treatment of mice with a neutralizing anti-cytokine antibody cocktail transiently improved early clinical and laboratory features of CRS. We discuss the predictive use of this model in the context of new anti-cytokine strategies to treat human CRS.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos/imunologia , Complexo CD3/antagonistas & inibidores , Síndrome da Liberação de Citocina/etiologia , Síndrome da Liberação de Citocina/metabolismo , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Animais , Anticorpos/efeitos adversos , Anticorpos Monoclonais/uso terapêutico , Síndrome da Liberação de Citocina/diagnóstico , Síndrome da Liberação de Citocina/tratamento farmacológico , Citocinas/sangue , Modelos Animais de Doenças , Quimioterapia Combinada , Mediadores da Inflamação/sangue , Mediadores da Inflamação/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Fenótipo , Índice de Gravidade de Doença , Linfócitos T/imunologia , Linfócitos T/metabolismo , Resultado do Tratamento
2.
Blood ; 134(17): 1430-1440, 2019 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-31383641

RESUMO

Antibodies that bind CD47 on tumor cells and prevent interaction with SIRPα on phagocytes are active against multiple cancer types including T-cell lymphoma (TCL). Here we demonstrate that surface CD47 is heterogeneously expressed across primary TCLs, whereas major histocompatibility complex (MHC) class I, which can also suppress phagocytosis, is ubiquitous. Multiple monoclonal antibodies (mAbs) that block CD47-SIRPα interaction promoted phagocytosis of TCL cells, which was enhanced by cotreatment with antibodies targeting MHC class I. Expression levels of surface CD47 and genes that modulate CD47 pyroglutamation did not correlate with the extent of phagocytosis induced by CD47 blockade in TCL lines. In vivo treatment of multiple human TCL patient-derived xenografts or an immunocompetent murine TCL model with a short course of anti-CD47 mAb markedly reduced lymphoma burden and extended survival. Depletion of macrophages reduced efficacy in vivo, whereas depletion of neutrophils had no effect. F(ab')2-only fragments of anti-CD47 antibodies failed to induce phagocytosis by human macrophages, indicating a requirement for Fc-Fcγ receptor interactions. In contrast, F(ab')2-only fragments increased phagocytosis by murine macrophages independent of SLAMF7-Mac-1 interaction. Full-length anti-CD47 mAbs also induced phagocytosis by Fcγ receptor-deficient murine macrophages. An immunoglobulin G1 anti-CD47 mAb induced phagocytosis and natural killer cell-mediated cytotoxicity of TCL cells that was augmented by cotreatment with mogamulizumab, an anti-CCR4 mAb, or a mAb blocking MHC class I. These studies help explain the disparate activity of monotherapy with agents that block CD47 in murine models compared with patients. They also have direct translational implications for the deployment of anti-CD47 mAbs alone or in combination.


Assuntos
Antígenos de Diferenciação/imunologia , Antineoplásicos Imunológicos/farmacologia , Antígeno CD47/imunologia , Linfoma de Células T/tratamento farmacológico , Receptores de IgG/imunologia , Receptores Imunológicos/imunologia , Animais , Antineoplásicos Imunológicos/uso terapêutico , Antígeno CD47/antagonistas & inibidores , Linhagem Celular Tumoral , Humanos , Linfoma de Células T/imunologia , Linfoma de Células T/patologia , Camundongos , Receptores Fc/imunologia
3.
J Immunol ; 192(4): 1641-50, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24442438

RESUMO

B cells play a major role in the pathogenesis of many autoimmune disorders, including rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, and type I diabetes mellitus, as indicated by the efficacy of B cell-targeted therapies in these diseases. Therapeutic effects of the most commonly used B cell-targeted therapy, anti-CD20 mAb, are contingent upon long-term depletion of peripheral B cells. In this article, we describe an alternative approach involving the targeting of CD79, the transducer subunit of the B cell AgR. Unlike anti-CD20 mAbs, the protective effects of CD79-targeted mAbs do not require cell depletion; rather, they act by inducing an anergic-like state. Thus, we describe a novel B cell-targeted approach predicated on the induction of B cell anergy.


Assuntos
Doenças Autoimunes/prevenção & controle , Linfócitos B/imunologia , Antígenos CD79/imunologia , Anergia Clonal/imunologia , Animais , Anticorpos Monoclonais/imunologia , Autoimunidade/imunologia , Feminino , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Depleção Linfocítica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout
4.
Exp Hematol Oncol ; 11(1): 26, 2022 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-35538512

RESUMO

BACKGROUND: CD47/SIRPα axis is recognized as an innate immune checkpoint and emerging clinical data validate the interest of interrupting this pathway in cancer, particularly in hematological malignancies. In preclinical models, CD47/SIRPα blocking agents have been shown to mobilize phagocytic cells and trigger adaptive immune responses to eliminate tumors. Here, we describe the mechanisms afforded by a CD47xCD19 bispecific antibody (NI-1701) at controlling tumor growth in a mouse xenograft B-cell lymphoma model. METHODS: The contribution of immune effector cell subsets behind the antitumor activity of NI-1701 was investigated using flow cytometry, transcriptomic analysis, and in vivo immune-cell depletion experiments. RESULTS: We showed that NI-1701 treatment transformed the tumor microenvironment (TME) into a more anti-tumorigenic state with increased NK cells, monocytes, dendritic cells (DC) and MHCIIhi tumor-associated macrophages (TAMs) and decreased granulocytic myeloid-derived suppressor cells. Notably, molecular analysis of isolated tumor-infiltrating leukocytes following NI-1701 administration revealed an upregulation of genes linked to immune activation, including IFNγ and IL-12b. Moreover, TAM-mediated phagocytosis of lymphoma tumor cells was enhanced in the TME in the presence of NI-1701, highlighting the role of macrophages in tumor control. In vivo cell depletion experiments demonstrated that both macrophages and NK cells contribute to the antitumor activity. In addition, NI-1701 enhanced dendritic cell-mediated phagocytosis of tumor cells in vitro, resulting in an increased cross-priming of tumor-specific CD8 T cells. CONCLUSIONS: The study described the mechanisms afforded by the CD47xCD19 bispecific antibody, NI-1701, at controlling tumor growth in lymphoma mouse model. NI-1701 is currently being evaluated in a Phase I clinical trial for the treatment of refractory or relapsed B-cell lymphoma (NCT04806035).

5.
MAbs ; 12(1): 1739408, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32191151

RESUMO

Mesothelin (MSLN) is a cell surface glycoprotein overexpressed in several solid malignancies, including gastric, lung, mesothelioma, pancreatic and ovarian cancers. While several MSLN-targeting therapeutic approaches are in development, only limited efficacy has been achieved in patients. A potential shortcoming of several described antibody-based approaches is that they target the membrane distal region of MSLN and, additionally, are known to be handicapped by the high levels of circulating soluble MSLN in patients. We show here, using monoclonal antibodies (mAbs) targeting different MSLN-spanning epitopes, that the membrane-proximal region resulted in more efficient killing of MSLN-positive tumor cells in antibody-dependent cell-mediated cytotoxicity (ADCC) assays. Surprisingly, no augmented killing was observed in antibody-dependent cellular phagocytosis (ADCP) by mAbs targeting this membrane-proximal region. To further increase the ADCP potential, we, therefore, generated bispecific antibodies (bsAbs) coupling a high-affinity MSLN binding arm to a blocking CD47 arm. Here, targeting the membrane-proximal domain of MSLN demonstrated enhanced ADCP activity compared to membrane-distal domains when the bsAbs were used in in vitro phagocytosis killing assays. Importantly, the superior anti-tumor activity was also translated in xenograft tumor models. Furthermore, we show that the bsAb approach targeting the membrane-proximal epitope of MSLN optimized ADCC activity by augmenting FcγR-IIIA activation and enhanced ADCP via a more efficient blockade of the CD47/SIRPα axis.


Assuntos
Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais/imunologia , Antineoplásicos/imunologia , Antígeno CD47/imunologia , Proteínas Ligadas por GPI/imunologia , Animais , Anticorpos Biespecíficos/farmacologia , Anticorpos Monoclonais/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antineoplásicos/farmacologia , Epitopos/imunologia , Humanos , Imunoterapia/métodos , Mesotelina , Camundongos , Fagocitose/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Antibodies (Basel) ; 7(3)2018 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-31544881

RESUMO

Bispecific antibodies (bsAbs) are often composed of several polypeptide chains that have to be expressed adequately to enable optimal assembly and yield of the bsAb. κλ bodies are a bispecific format with a native IgG structure, composed of two different light chains that pair with a common heavy chain. Introduction of non-optimal codons into the sequence of a particular polypeptide is an effective strategy for down modulating its expression. Here we applied this strategy but restricted the modification of the codon content to the constant domain of one light chain. This approach facilitates parallel optimization of several bsAbs by using the same modified constant domains. Partial sequence de-optimization reduced expression of the targeted polypeptide. Stable cell pools could be isolated displaying increased bispecific antibody titers as well as changes in the abundance of undesired by-products that require elimination during downstream processing. Thus, modulating the relative expression of polypeptides can have a significant impact on bsAb titer and product related impurities; which are important factors for large scale manufacturing for clinical supply.

7.
MAbs ; 9(2): 231-239, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28001485

RESUMO

When production of bispecific antibodies requires the co-expression and assembly of three or four polypeptide chains, low expression of one chain can significantly limit assembly and yield. κλ bodies, fully human bispecific antibodies with native IgG structure, are composed of a common heavy chain and two different light chains, one kappa and one lambda. No engineering is applied to force pairing of the chains, thus both monospecific and bispecific antibodies are secreted in the supernatant. In this context, stoichiometric expression of the two light chains allows for maximal assembly of the bispecific antibody. In this study, we selected a κλ body with suboptimal characteristics due to low kappa chain expression. Codon optimization to increase expression of the kappa chain did not improve bispecific yield. Surprisingly, progressive introduction of non-optimal codons into the sequence of the lambda chain resulted in lowering its expression for an optimal tuning of the relative distribution of monospecific and bispecific antibodies. This codon de-optimization led to doubling of the κλ body yield. These results indicate that assembly of different proteins into a recombinant complex is an interconnected process and that reducing the expression of one polypeptide can actually increase the overall yield.


Assuntos
Anticorpos Biespecíficos/biossíntese , Engenharia de Proteínas/métodos , Animais , Códon , Humanos , Imunoglobulina G/biossíntese , Cadeias kappa de Imunoglobulina/biossíntese , Cadeias kappa de Imunoglobulina/genética , Cadeias lambda de Imunoglobulina/biossíntese , Cadeias lambda de Imunoglobulina/genética
8.
J Mol Biol ; 427(16): 2647-62, 2015 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-26013163

RESUMO

Hu 15C1 is a potent anti-human Toll-like receptor 4 (TLR4) neutralizing antibody. To better understand the molecular basis of its biological activity, we used a multidisciplinary approach to generate an accurate model of the Hu 15C1-TLR4 complex. By combining site-directed mutagenesis, in vitro antibody evolution, affinity measurements and X-ray crystallography of Fab fragments, we identified key interactions across the Hu 15C1-TLR4 interface. These contact points were used as restraints to predict the structure of the Fab region of Hu 15C1 bound to TLR4 using computational molecular docking. This model was further evaluated and validated by additional site-directed mutagenesis studies. The predicted structure of the Hu 15C1-TLR4 complex indicates that the antibody antagonizes the receptor dimerization necessary for its activation. This study exemplifies how iterative cycles of antibody engineering can facilitate the discovery of components of antibody-target interactions.


Assuntos
Anticorpos Neutralizantes/imunologia , Complexo Antígeno-Anticorpo/ultraestrutura , Sítios de Ligação de Anticorpos/imunologia , Fragmentos Fab das Imunoglobulinas/ultraestrutura , Receptor 4 Toll-Like/imunologia , Sequência de Aminoácidos , Animais , Complexo Antígeno-Anticorpo/imunologia , Células CHO , Linhagem Celular , Técnicas de Visualização da Superfície Celular , Simulação por Computador , Cricetinae , Cricetulus , Cristalografia por Raios X , Ensaio de Imunoadsorção Enzimática , Epitopos/imunologia , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Modelos Moleculares , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação Proteica , Alinhamento de Sequência , Especificidade da Espécie , Ressonância de Plasmônio de Superfície
9.
Nat Commun ; 6: 6113, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25672245

RESUMO

Bispecific antibodies enable unique therapeutic approaches but it remains a challenge to produce them at the industrial scale, and the modifications introduced to achieve bispecificity often have an impact on stability and risk of immunogenicity. Here we describe a fully human bispecific IgG devoid of any modification, which can be produced at the industrial scale, using a platform process. This format, referred to as a κλ-body, is assembled by co-expressing one heavy chain and two different light chains, one κ and one λ. Using ten different targets, we demonstrate that light chains can play a dominant role in mediating specificity and high affinity. The κλ-bodies support multiple modes of action, and their stability and pharmacokinetic properties are indistinguishable from therapeutic antibodies. Thus, the κλ-body represents a unique, fully human format that exploits light-chain variable domains for antigen binding and light-chain constant domains for robust downstream processing, to realize the potential of bispecific antibodies.


Assuntos
Anticorpos Biespecíficos/isolamento & purificação , Imunoglobulina G/isolamento & purificação , Cadeias Pesadas de Imunoglobulinas/isolamento & purificação , Engenharia de Proteínas/métodos , Anticorpos Monoclonais/metabolismo , Cromatografia Líquida de Alta Pressão , Humanos , Cadeias Leves de Imunoglobulina/metabolismo , Cadeias kappa de Imunoglobulina/metabolismo , Testes de Neutralização , Biblioteca de Peptídeos , Linfócitos T/imunologia
10.
Cell Host Microbe ; 6(4): 354-66, 2009 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-19837375

RESUMO

Autophagy is a highly regulated self-degradative mechanism required at a basal level for intracellular clearance and recycling of cytoplasmic contents. Upon intracellular pathogen invasion, autophagy can be induced as an innate immune mechanism to control infection. Nevertheless, pathogens have developed strategies to avoid or hijack autophagy for their own benefit. The molecular pathways inducing autophagy in response to infection remain poorly documented. We report here that the engagement of CD46, a ubiquitous human surface receptor able to bind several different pathogens, is sufficient to induce autophagy. CD46-Cyt-1, one of the two C-terminal splice variants of CD46, is linked to the autophagosome formation complex VPS34/Beclin1 via its interaction with the scaffold protein GOPC. Measles virus and group A Streptococcus, two CD46-binding pathogens, induce autophagy through a CD46-Cyt-1/GOPC pathway. Thus, upon microorganism recognition, a cell surface pathogen receptor can directly trigger autophagy, a critical step to control infection.


Assuntos
Autofagia , Vírus do Sarampo/imunologia , Proteína Cofatora de Membrana/imunologia , Streptococcus pyogenes/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Proteínas de Transporte/metabolismo , Proteínas da Matriz do Complexo de Golgi , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Fosfatidilinositol 3-Quinases/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas
11.
Virology ; 375(2): 452-63, 2008 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-18358512

RESUMO

Caprine arthritis-encephalitis virus (CAEV) is a lentivirus that causes natural inflammatory disease in goats, with chronic lesions in several different organs. CAEV infection of in vitro cultured cells is accompanied by apoptosis, but the involvement of the intrinsic and extrinsic pathways has not previously been elucidated. We have studied the activation of caspases-3, -8 and -9 by fluorescent assays in various goat cells infected in vitro by CAEV, and the effects of transfected dominant negative variants of theses caspases, to show that CAEV-associated apoptosis depends on activation of caspases-3 and -9, but not -8. A simultaneous disruption of mitochondrial membrane potential indicates an involvement of mitochondrial pathway.


Assuntos
Apoptose , Vírus da Artrite-Encefalite Caprina/fisiologia , Caspase 3/metabolismo , Caspase 9/metabolismo , Infecções por Lentivirus/metabolismo , Infecções por Lentivirus/virologia , Animais , Caspase 8/metabolismo , Células Cultivadas , Cabras , Macrófagos , Mitocôndrias/metabolismo , Transdução de Sinais , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA