Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Autoimmun ; 95: 1-14, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30446251

RESUMO

Susceptibility to multiple autoimmune diseases is associated with common gene polymorphisms influencing IL-2 signaling and Treg function, making Treg-specific expansion by IL-2 a compelling therapeutic approach to treatment. As an in vivo IL-2 half-life enhancer we used a non-targeted, effector-function-silent human IgG1 as a fusion protein. An IL-2 mutein (N88D) with reduced binding to the intermediate affinity IL-2Rßγ receptor was engineered with a stoichiometry of two IL-2N88D molecules per IgG, i.e. IgG-(IL-2N88D)2. The reduced affinity of IgG-(IL-2N88D)2 for the IL-2Rßγ receptor resulted in a Treg-selective molecule in human whole blood pSTAT5 assays. Treatment of cynomolgus monkeys with single low doses of IgG-(IL-2N88D)2 induced sustained preferential activation of Tregs accompanied by a corresponding 10-14-fold increase in CD4+ and CD8+ CD25+FOXP3+ Tregs; conditions that had no effect on CD4+ or CD8+ memory effector T cells. The expanded cynomolgus Tregs had demethylated FOXP3 and CTLA4 epigenetic signatures characteristic of functionally suppressive cells. Humanized mice had similar selective in vivo responses; IgG-(IL-2N88D)2 increased Tregs while wild-type IgG-IL-2 increased NK cells in addition to Tregs. The expanded human Tregs had demethylated FOXP3 and CTLA4 signatures and were immunosuppressive. These results describe a next-generation immunotherapy using a long-lived and Treg-selective IL-2 that activates and expands functional Tregsin vivo. Patients should benefit from restored immune homeostasis in a personalized fashion to the extent that their autoimmune disease condition dictates opening up the possibility for remissions and cures.


Assuntos
Doenças Autoimunes/terapia , Imunoglobulina G/imunologia , Imunoterapia/métodos , Interleucina-2/imunologia , Linfotoxina-alfa/imunologia , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Sítios de Ligação , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Antígeno CTLA-4/genética , Antígeno CTLA-4/imunologia , Proliferação de Células , Metilação de DNA/efeitos dos fármacos , Modelos Animais de Doenças , Epigênese Genética , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Humanos , Imunoglobulina G/administração & dosagem , Imunoglobulina G/química , Imunoglobulina G/genética , Interleucina-2/administração & dosagem , Interleucina-2/química , Interleucina-2/genética , Subunidade beta de Receptor de Interleucina-2/genética , Subunidade beta de Receptor de Interleucina-2/imunologia , Ativação Linfocitária/efeitos dos fármacos , Linfotoxina-alfa/administração & dosagem , Linfotoxina-alfa/química , Linfotoxina-alfa/genética , Macaca fascicularis , Masculino , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Estrutura Secundária de Proteína , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/imunologia , Transdução de Sinais , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia
2.
Sci Transl Med ; 10(461)2018 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-30282693

RESUMO

T cell bispecific antibodies (TCBs) are engineered molecules that include, within a single entity, binding sites to the T cell receptor and to tumor-associated or tumor-specific antigens. The receptor tyrosine kinase HER2 is a tumor-associated antigen in ~25% of breast cancers. TCBs targeting HER2 may result in severe toxicities, likely due to the expression of HER2 in normal epithelia. About 40% of HER2-positive tumors express p95HER2, a carboxyl-terminal fragment of HER2. Using specific antibodies, here, we show that p95HER2 is not expressed in normal tissues. We describe the development of p95HER2-TCB and show that it has a potent antitumor effect on p95HER2-expressing breast primary cancers and brain lesions. In contrast with a TCB targeting HER2, p95HER2-TCB has no effect on nontransformed cells that do not overexpress HER2. These data pave the way for the safe treatment of a subgroup of HER2-positive tumors by targeting a tumor-specific antigen.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Receptor ErbB-2/imunologia , Linfócitos T/imunologia , Animais , Neoplasias da Mama/patologia , Complexo CD3/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Exp Med ; 215(3): 859-876, 2018 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-29436396

RESUMO

Depletion of immunosuppressive tumor-associated macrophages (TAMs) or reprogramming toward a proinflammatory activation state represent different strategies to therapeutically target this abundant myeloid population. In this study, we report that inhibition of colony-stimulating factor-1 receptor (CSF-1R) signaling sensitizes TAMs to profound and rapid reprogramming in the presence of a CD40 agonist before their depletion. Despite the short-lived nature of macrophage hyperactivation, combined CSF-1R+CD40 stimulation of macrophages is sufficient to create a proinflammatory tumor milieu that reinvigorates an effective T cell response in transplanted tumors that are either responsive or insensitive to immune checkpoint blockade. The central role of macrophages in regulating preexisting immunity is substantiated by depletion experiments, transcriptome analysis of ex vivo sorted TAMs, and gene expression profiling of whole tumor lysates at an early treatment time point. This approach enabled the identification of specific combination-induced changes among the pleiotropic activation spectrum of the CD40 agonist. In patients, CD40 expression on human TAMs was detected in mesothelioma and colorectal adenocarcinoma.


Assuntos
Imunidade , Macrófagos/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Animais , Antígenos CD40/agonistas , Antígenos CD40/metabolismo , Linfócitos T CD8-Positivos/imunologia , Feminino , Humanos , Inflamação/patologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fenótipo , Receptor de Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo
4.
Oncoimmunology ; 6(3): e1277306, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28405498

RESUMO

We developed cergutuzumab amunaleukin (CEA-IL2v, RG7813), a novel monomeric CEA-targeted immunocytokine, that comprises a single IL-2 variant (IL2v) moiety with abolished CD25 binding, fused to the C-terminus of a high affinity, bivalent carcinoembryonic antigen (CEA)-specific antibody devoid of Fc-mediated effector functions. Its molecular design aims to (i) avoid preferential activation of regulatory T-cells vs. immune effector cells by removing CD25 binding; (ii) increase the therapeutic index of IL-2 therapy by (a) preferential retention at the tumor by having a lower dissociation rate from CEA-expressing cancer cells vs. IL-2R-expressing cells, (b) avoiding any FcγR-binding and Fc effector functions and (c) reduced binding to endothelial cells expressing CD25; and (iii) improve the pharmacokinetics, and thus convenience of administration, of IL-2. The crystal structure of the IL2v-IL-2Rßγ complex was determined and CEA-IL2v activity was assessed using human immune effector cells. Tumor targeting was investigated in tumor-bearing mice using 89Zr-labeled CEA-IL2v. Efficacy studies were performed in (a) syngeneic mouse models as monotherapy and combined with anti-PD-L1, and in (b) xenograft mouse models in combination with ADCC-mediating antibodies. CEA-IL2v binds to CEA with pM avidity but not to CD25, and consequently did not preferentially activate Tregs. In vivo, CEA-IL2v demonstrated superior pharmacokinetics and tumor targeting compared with a wild-type IL-2-based CEA immunocytokine (CEA-IL2wt). CEA-IL2v strongly expanded NK and CD8+ T cells, skewing the CD8+:CD4+ ratio toward CD8+ T cells both in the periphery and in the tumor, and mediated single agent efficacy in syngeneic MC38-CEA and PancO2-CEA models. Combination with trastuzumab, cetuximab and imgatuzumab, all of human IgG1 isotype, resulted in superior efficacy compared with the monotherapies alone. Combined with anti-PD-L1, CEA-IL2v mediated superior efficacy over the respective monotherapies, and over the combination with an untargeted control immunocytokine. These preclinical data support the ongoing clinical investigation of the cergutuzumab amunaleukin immunocytokine with abolished CD25 binding for the treatment of CEA-positive solid tumors in combination with PD-L1 checkpoint blockade and ADCC competent antibodies.

5.
Clin Cancer Res ; 22(13): 3286-97, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-26861458

RESUMO

PURPOSE: CEA TCB is a novel IgG-based T-cell bispecific (TCB) antibody for the treatment of CEA-expressing solid tumors currently in phase I clinical trials (NCT02324257). Its format incorporates bivalent binding to CEA, a head-to-tail fusion of CEA- and CD3e-binding Fab domains and an engineered Fc region with completely abolished binding to FcγRs and C1q. The study provides novel mechanistic insights into the activity and mode of action of CEA TCB. EXPERIMENTAL DESIGN: CEA TCB activity was characterized on 110 cell lines in vitro and in xenograft tumor models in vivo using NOG mice engrafted with human peripheral blood mononuclear cells. RESULTS: Simultaneous binding of CEA TCB to tumor and T cells leads to formation of immunologic synapses, T-cell activation, secretion of cytotoxic granules, and tumor cell lysis. CEA TCB activity strongly correlates with CEA expression, with higher potency observed in highly CEA-expressing tumor cells and a threshold of approximately 10,000 CEA-binding sites/cell, which allows distinguishing between high- and low-CEA-expressing tumor and primary epithelial cells, respectively. Genetic factors do not affect CEA TCB activity confirming that CEA expression level is the strongest predictor of CEA TCB activity. In vivo, CEA TCB induces regression of CEA-expressing xenograft tumors with variable amounts of immune cell infiltrate, leads to increased frequency of activated T cells, and converts PD-L1 negative into PD-L1-positive tumors. CONCLUSIONS: CEA TCB is a novel generation TCB displaying potent antitumor activity; it is efficacious in poorly infiltrated tumors where it increases T-cell infiltration and generates a highly inflamed tumor microenvironment. Clin Cancer Res; 22(13); 3286-97. ©2016 AACR.


Assuntos
Anticorpos Biespecíficos/farmacologia , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Antígeno Carcinoembrionário/imunologia , Imunoterapia/métodos , Neoplasias/terapia , Linfócitos T/imunologia , Animais , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Antineoplásicos/imunologia , Sítios de Ligação/imunologia , Complexo CD3/imunologia , Linhagem Celular Tumoral , Feminino , Humanos , Ativação Linfocitária/imunologia , Camundongos , Receptores Fc/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Autoimmun ; 56: 66-80, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25457307

RESUMO

Regulatory T cells (Tregs) expressing FOXP3 are essential for the maintenance of self-tolerance and are deficient in many common autoimmune diseases. Immune tolerance is maintained in part by IL-2 and deficiencies in the IL-2 pathway cause reduced Treg function and an increased risk of autoimmunity. Recent studies expanding Tregs in vivo with low-dose IL-2 achieved major clinical successes highlighting the potential to optimize this pleiotropic cytokine for inflammatory and autoimmune disease indications. Here we compare the clinically approved IL-2 molecule, Proleukin, with two engineered IL-2 molecules with long half-lives owing to their fusion in monovalent and bivalent stoichiometry to a non-FcRγ binding human IgG1. Using nonhuman primates, we demonstrate that single ultra-low doses of IL-2 fusion proteins induce a prolonged state of in vivo activation that increases Tregs for an extended period of time similar to multiple-dose Proleukin. One of the common pleiotropic effects of high dose IL-2 treatment, eosinophilia, is eliminated at doses of the IL-2 fusion proteins that greatly expand Tregs. The long half-lives of the IL-2 fusion proteins facilitated a detailed characterization of an IL-2 dose response driving Treg expansion that correlates with increasingly sustained, suprathreshold pSTAT5a induction and subsequent sustained increases in the expression of CD25, FOXP3 and Ki-67 with retention of Treg-specific epigenetic signatures at FOXP3 and CTLA4.


Assuntos
Interleucina-2/metabolismo , Transdução de Sinais , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Biomarcadores/metabolismo , Antígeno CTLA-4/metabolismo , Relação Dose-Resposta a Droga , Eosinofilia/induzido quimicamente , Feminino , Fatores de Transcrição Forkhead/metabolismo , Humanos , Interleucina-2/análogos & derivados , Interleucina-2/farmacologia , Subunidade alfa de Receptor de Interleucina-2/deficiência , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Contagem de Linfócitos , Macaca fascicularis , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Fosforilação/efeitos dos fármacos , Ligação Proteica , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes/farmacologia , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos
7.
Anal Biochem ; 376(2): 163-72, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18342613

RESUMO

This study illustrates the application of asymmetrical flow field-flow fractionation (AF4) and light scattering analysis during the development of a gene delivery vehicle based on virus-like particles (VLPs) derived from the human polyoma JC virus. The analytical system was created by connecting an AF4 apparatus to the following detectors: diode array, fluorescence, multiangle light scattering, dynamic light scattering, and refractometer. From a single analysis, the molar mass, root mean square and hydrodynamic radii, composition, and purity of the sample could be determined. The VLPs were purified from baculovirus-infected Sf158 insect cells overexpressing the recombinant VP1 protein using weak anion exchange chromatography. The VLPs were dissociated to VP1 pentamers, and the contaminating DNA and proteins were removed using strong anion exchange chromatography. The gene delivery vehicle was created by reassembling the VP1 pentamers in the presence of the desired DNA. The newly formed VLPs encapsulated the DNA and were shown to be capable of delivering the gene of interest to target cells where it was translated into protein. This paper describes the scalable process that was derived to produce the VLPs and demonstrates how the AF4-based analytical characterization was indispensable during the development process.


Assuntos
DNA/genética , Fracionamento por Campo e Fluxo/métodos , Técnicas de Transferência de Genes , Polyomavirus/genética , Espalhamento de Radiação , Animais , Linhagem Celular , Humanos , Luz , Proteínas Recombinantes/metabolismo , Spodoptera
8.
J Interferon Cytokine Res ; 26(10): 699-705, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17032164

RESUMO

Recombinant interferon-beta (IFN-beta) protein is used successfully for the treatment of multiple sclerosis (MS). Gene therapy might be an alternative approach to overcome drawbacks occurring with IFN-beta protein therapy. A critical issue in developing a new approach is detection of biologically active IFN-beta in preclinical models. The goal of the present study was to determine if Mx1 and IP-10, which are known to be activated after IFN-beta treatment in humans, can be used as biomarkers in mice. In three in vivo experiments, the correlation between different methods of murine IFN-beta (MuIFN-beta) delivery and biomarker induction was studied: (1) bolus protein delivery by intravenous (i.v.) or intramuscular (i.m.) injection, (2) gene-based delivery of IFN- beta by i.m. injection of plasmid DNA, followed by electroporation, and (3) gene-based delivery of IFN-beta by i.m. injection of adenovirus-associated type 1 (AAV1). Short-term induction of Mx1 mRNA and IP-10 was observed after treatment with bolus MuIFN-beta protein. Long-term induction of both biomarkers was observed after IFN-beta plasmid DNA delivery or when AAV1 was used as the vector. The experiments demonstrate that gene-based delivery provides sustained levels of IFN-beta compared with bolus protein injection and that Mx1 RNA and IP-10 can be used to monitor biologically active circulating plasma MuIFN-beta protein in mice.


Assuntos
Quimiocinas CXC/biossíntese , Proteínas de Ligação ao GTP/biossíntese , Técnicas de Transferência de Genes , Interferon beta/genética , Animais , Biomarcadores/metabolismo , Quimiocina CXCL10 , Quimiocinas CXC/genética , Dependovirus/genética , Eletroporação , Proteínas de Ligação ao GTP/genética , Vetores Genéticos , Injeções , Interferon beta/administração & dosagem , Interferon beta/farmacologia , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Resistência a Myxovirus , Plasmídeos/genética , RNA Mensageiro/metabolismo
9.
Curr Opin Mol Ther ; 6(3): 302-7, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15264433

RESUMO

The antiretroviral treatment of HIV infection has expanded tremendously in the last few years. This trend is mainly a result of new assays, genetic advances and recombinant DNA technologies providing new insights into HIV virology, pathology and resistance mechanisms. Since 1995, the use of highly active antiretroviral therapy (HAART) as a combination of substances directed against various steps in the viral life cycle, has led to significant decreases in the morbidity and mortality associated with HIV infections. The ability to quantify viral load and to perform sequence analyses represent valuable tools both for understanding the pathogenic actions of the virus and for clinical drug monitoring of HIV-infected patients. Laboratory tests have been developed and validated to help predict which antiviral substances may be more effective to control viral replication in a given patient. Genotypic and phenotypic assays have been developed to assess HIV antiviral resistance. The ability to predict drug response from a certain genotypic or phenotypic setting with respect to drug absorption, drug metabolism and drug elimination is continually evolving. This may lead to significant changes in drug plasma concentrations and may affect the efficacy and toxicity of antivirals. More recently, drug/drug interactions and mutation/drug correlations have been discovered. However, the optimal usage of pharmacogenetic and pharmacogenomic tools in the clinic still remains to be defined. This review summarizes mechanisms of drug bioavailability with respect to pharmacogenomics and discusses the impact and clinical benefit of 'personalized' HIV therapy.


Assuntos
Síndrome da Imunodeficiência Adquirida/tratamento farmacológico , Terapia Antirretroviral de Alta Atividade , Antivirais/uso terapêutico , Farmacorresistência Viral/genética , HIV/fisiologia , Farmacogenética , Síndrome da Imunodeficiência Adquirida/genética , Síndrome da Imunodeficiência Adquirida/patologia , Terapia Antirretroviral de Alta Atividade/tendências , Feminino , HIV/efeitos dos fármacos , Humanos , Masculino , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
10.
Curr Opin Mol Ther ; 5(5): 524-8, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14601522

RESUMO

A major challenge in the field of gene therapy is the development of new carrier/delivery systems that lack the disadvantages of current transfer systems. In the past, some time has been spent developing such modified or alternative vectors. A new candidate is represented by virus-like particles (VLPs). It has been shown that recombinant expression of the major structural proteins of many viruses leads to the formation of VLPs. Such VLPs exhibit morphology similar to the empty capsids of the virus from which they are derived. VLPs are non-infectious, have a similar tropism to the natural virus, and show comparable cellular uptake and intracellular trafficking. Since its discovery, VLP technology has gained importance in biomedical research. Although most investigations into VLP technology have dealt with vaccine development, some research groups have demonstrated that VLPs could also represent a useful gene therapy delivery system. This review will focus on studies performed with VLPs from members of the Papillomaviridae and Polyomaviridae families.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/metabolismo , Vírion/genética , Animais , DNA Viral/metabolismo , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Humanos , Papillomaviridae/genética , Papillomaviridae/metabolismo , Polyomaviridae/genética , Polyomaviridae/metabolismo , Vírion/imunologia , Vírion/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA