Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Rheumatology (Oxford) ; 62(12): 4006-4011, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37335864

RESUMEN

OBJECTIVE: Immune-mediated necrotizing myopathies (IMNMs) are severe forms of myositis often associated with pathogenic anti-3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) autoantibodies (aAbs). Efgartigimod is an engineered human IgG1 Fc fragment that antagonizes the neonatal Fc receptor (FcRn), thereby preventing recycling and promoting lysosomal degradation of IgG, including aAbs. We evaluated the therapeutic effects of IgG reduction by efgartigimod in a humanized murine model of IMNM. METHODS: Disease was induced in C5-deficient (C5def) or Rag2-deficient (Rag2-/-) mice receiving co-injections of anti-HMGCR+ IgG from an IMNM patient and human complement. C5def mice were treated in a preventive setting with s.c. injections of efgartigimod and Rag2-/- mice in a curative setting after disease was induced by anti-HMGCR+ IgG injections. Anti-HMGCR aAbs levels were monitored in mouse serum and muscle tissue. Histological analysis was performed on muscle sections. Muscle force was assessed by grip test or measurement of gastrocnemius strength upon electrostimulation. RESULTS: Administration of efgartigimod rapidly reduced total IgG levels, including the level of pathogenic anti-HMGCR aAbs, in both serum (P < 0.0001) and muscle (P < 0.001). In the preventive setting, efgartigimod prevented myofibre necrosis (P < 0.05), thus precluding loss of muscle strength (P < 0.05). In the therapeutic setting, efgartigimod prevented further necrosis and allowed muscle fibre regeneration (P < 0.05). Hence, muscle strength returned to normal (P < 0.01). CONCLUSION: Efgartigimod reduces circulating IgG levels, including pathogenic anti-HMGCR+ IgG aAbs, in a humanized mouse model of IMNM, preventing further necrosis and allowing muscle fibre regeneration. These results support investigating the therapeutic efficacy of efgartigimod through a clinical trial in IMNM patients.


Asunto(s)
Enfermedades Autoinmunes , Enfermedades Musculares , Miositis , Humanos , Animales , Ratones , Modelos Animales de Enfermedad , Músculo Esquelético/patología , Autoanticuerpos , Hidroximetilglutaril-CoA Reductasas , Inmunoglobulina G , Necrosis
2.
Oncoimmunology ; 11(1): 2144669, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36387055

RESUMEN

Combining immunogenic cell death-inducing chemotherapies and PD-1 blockade can generate remarkable tumor responses. It is now well established that TGF-ß1 signaling is a major component of treatment resistance and contributes to the cancer-related immunosuppressive microenvironment. However, whether TGF-ß1 remains an obstacle to immune checkpoint inhibitor efficacy when immunotherapy is combined with chemotherapy is still to be determined. Several syngeneic murine models were used to investigate the role of TGF-ß1 neutralization on the combinations of immunogenic chemotherapy (FOLFOX: 5-fluorouracil and oxaliplatin) and anti-PD-1. Cancer-associated fibroblasts (CAF) and immune cells were isolated from CT26 and PancOH7 tumor-bearing mice treated with FOLFOX, anti-PD-1 ± anti-TGF-ß1 for bulk and single cell RNA sequencing and characterization. We showed that TGF-ß1 neutralization promotes the therapeutic efficacy of FOLFOX and anti-PD-1 combination and induces the recruitment of antigen-specific CD8+ T cells into the tumor. TGF-ß1 neutralization is required in addition to chemo-immunotherapy to promote inflammatory CAF infiltration, a chemokine production switch in CAF leading to decreased CXCL14 and increased CXCL9/10 production and subsequent antigen-specific T cell recruitment. The immune-suppressive effect of TGF-ß1 involves an epigenetic mechanism with chromatin remodeling of CXCL9 and CXCL10 promoters within CAF DNA in a G9a and EZH2-dependent fashion. Our results strengthen the role of TGF-ß1 in the organization of a tumor microenvironment enriched in myofibroblasts where chromatin remodeling prevents CXCL9/10 production and limits the efficacy of chemo-immunotherapy.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias , Ratones , Animales , Fibroblastos Asociados al Cáncer/patología , Linfocitos T CD8-positivos , Inmunoterapia/métodos , Quimiocinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
4.
Nat Commun ; 11(1): 4545, 2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917858

RESUMEN

TGF-ß1, ß2 and ß3 bind a common receptor to exert vastly diverse effects in cancer, supporting either tumor progression by favoring metastases and inhibiting anti-tumor immunity, or tumor suppression by inhibiting malignant cell proliferation. Global TGF-ß inhibition thus bears the risk of undesired tumor-promoting effects. We show that selective blockade of TGF-ß1 production by Tregs with antibodies against GARP:TGF-ß1 complexes induces regressions of mouse tumors otherwise resistant to anti-PD-1 immunotherapy. Effects of combined GARP:TGF-ß1/PD-1 blockade are immune-mediated, do not require FcγR-dependent functions and increase effector functions of anti-tumor CD8+ T cells without augmenting immune cell infiltration or depleting Tregs within tumors. We find GARP-expressing Tregs and evidence that they produce TGF-ß1 in one third of human melanoma metastases. Our results suggest that anti-GARP:TGF-ß1 mAbs, by selectively blocking a single TGF-ß isoform emanating from a restricted cellular source exerting tumor-promoting activity, may overcome resistance to PD-1/PD-L1 blockade in patients with cancer.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas de la Membrana/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral/trasplante , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/inmunología , Células HEK293 , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Neoplasias/inmunología , Neoplasias/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
5.
Mol Pharmacol ; 96(6): 753-764, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31481460

RESUMEN

Dysregulation of the chemokine system is implicated in a number of autoimmune and inflammatory diseases, as well as cancer. Modulation of chemokine receptor function is a very promising approach for therapeutic intervention. Despite interest from academic groups and pharmaceutical companies, there are currently few approved medicines targeting chemokine receptors. Monoclonal antibodies (mAbs) and antibody-based molecules have been successfully applied in the clinical therapy of cancer and represent a potential new class of therapeutics targeting chemokine receptors belonging to the class of G protein-coupled receptors (GPCRs). Besides conventional mAbs, single-domain antibodies and antibody scaffolds are also gaining attention as promising therapeutics. In this review, we provide an extensive overview of mAbs, single-domain antibodies, and other antibody fragments targeting CXCR4 and ACKR3, formerly referred to as CXCR7. We discuss their unique properties and advantages over small-molecule compounds, and also refer to the molecules in preclinical and clinical development. We focus on single-domain antibodies and scaffolds and their utilization in GPCR research. Additionally, structural analysis of antibody binding to CXCR4 is discussed. SIGNIFICANCE STATEMENT: Modulating the function of GPCRs, and particularly chemokine receptors, draws high interest. A comprehensive review is provided for monoclonal antibodies, antibody fragments, and variants directed at CXCR4 and ACKR3. Their advantageous functional properties, versatile applications as research tools, and use in the clinic are discussed.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/metabolismo , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Anticuerpos de Dominio Único/administración & dosificación , Anticuerpos de Dominio Único/metabolismo , Animales , Sistemas de Liberación de Medicamentos/métodos , Humanos , Receptores CXCR/antagonistas & inhibidores , Receptores CXCR4/antagonistas & inhibidores
6.
Biochem Pharmacol ; 158: 413-424, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30342023

RESUMEN

Upregulation of the chemokine receptor CXCR4 contributes to the progression and metastasis of both solid and hematological malignancies, rendering this receptor an attractive therapeutic target. Besides the only FDA-approved CXCR4 antagonist Plerixafor (AMD3100), multiple other classes of CXCR4-targeting molecules are under (pre-)clinical development. Nanobodies (Nb), small single variable domains of heavy-chain only antibodies from Camelids, have appeared to be ideal antibody-fragments for targeting a broad range of epitopes and cavities within GPCRs such as CXCR4. Compared to conventional antibodies, monovalent nanobodies show fast blood clearance and no effector functions. In order to further increase their binding affinities and to restore antibody-mediated effector functions, we have constructed three different bivalent nanobody Fc-fusion molecules (Nb-Fc), targeting distinct epitopes on CXCR4, via fusion of Nbs to a Fc domain of a human IgG1 antibody. Most Nb-Fc constructs show increased binding affinity and enhanced potency in CXCL12 displacement, inhibition of CXCL12-induced signaling and CXCR4-mediated HIV entry, when compared to their monovalent Nb counterparts. Moreover, Nb-Fc induced ADCC- and CDC-mediated cell-death of CXCR4-overexpressing CCRF-CEM leukemia cells and did not affect cells expressing low levels or no CXCR4. These highly potent CXCR4 Nb-Fc constructs with Fc-mediated effector functions are attractive molecules to therapeutically target CXCR4-overexpressing tumors.


Asunto(s)
Inhibidores de Fusión de VIH/administración & dosificación , Inmunoglobulina G/administración & dosificación , Receptores CXCR4/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Anticuerpos de Dominio Único/administración & dosificación , Animales , Células CHO , Cricetulus , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Inmunoglobulina G/química , Células Jurkat , Estructura Secundaria de Proteína , Receptores CXCR4/metabolismo , Transducción de Señal/fisiología , Anticuerpos de Dominio Único/química
7.
Biochem Pharmacol ; 158: 402-412, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30342024

RESUMEN

The chemokine receptor CXCR4 and its ligand CXCL12 contribute to a variety of human diseases, such as cancer. CXCR4 is also a major co-receptor facilitating HIV entry. Accordingly, CXCR4 is considered as an attractive therapeutic target. Drug side effects and poor pharmacokinetic properties have been major hurdles that have prevented the implementation of CXCR4-directed inhibitors in treatment regimes. We evaluated the activity of a new and promising class of biologics, namely CXCR4-targeting nanobodies, with the purpose of identifying nanobodies that would preferentially inhibit HIV infection, while minimally disturbing other CXCR4-related functions. All CXCR4-interacting nanobodies inhibited CXCL12 binding and receptor-mediated calcium mobilization with comparable relative potencies. Importantly, the anti-HIV-1 activity of the nanobodies did not always correlate with their ability to modulate CXCR4 signaling and function, indicating that the anti-HIV and anti-CXCR4 activity are not entirely overlapping and may be functionally separated. Three nanobodies with divergent activity profiles (VUN400, VUN401 and VUN402) were selected for in depth biological evaluation. While all three nanobodies demonstrated inhibitory activity against a wide range of HIV (X4) strains, VUN402 poorly blocked CXCL12-induced CXCR4 internalization, chemotaxis and changes in cell morphology. Each of these nanobodies recognized distinct, although partially overlapping epitopes on CXCR4, which might underlie their distinct activity profiles. Our results demonstrate the potential of CXCR4-targeting nanobody VUN402 as a novel lead and starting point for the development of a more potent and selective anti-HIV agent.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Inhibidores de Fusión de VIH/administración & dosificación , VIH-1/efectos de los fármacos , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/fisiología , Anticuerpos de Dominio Único/administración & dosificación , Animales , Camélidos del Nuevo Mundo , Relación Dosis-Respuesta a Droga , Inhibidores de Fusión de VIH/metabolismo , VIH-1/metabolismo , Humanos , Células Jurkat , Estructura Secundaria de Proteína , Ratas , Anticuerpos de Dominio Único/metabolismo
8.
Science ; 362(6417): 952-956, 2018 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-30361387

RESUMEN

Transforming growth factor-ß1 (TGF-ß1) is one of very few cytokines produced in a latent form, requiring activation to exert any of its vastly diverse effects on development, immunity, and cancer. Regulatory T cells (Tregs) suppress immune cells within close proximity by activating latent TGF-ß1 presented by GARP (glycoprotein A repetitions predominant) to integrin αVß8 on their surface. We solved the crystal structure of GARP:latent TGF-ß1 bound to an antibody that stabilizes the complex and blocks release of active TGF-ß1. This finding reveals how GARP exploits an unusual medley of interactions, including fold complementation by the amino terminus of TGF-ß1, to chaperone and orient the cytokine for binding and activation by αVß8. Thus, this work further elucidates the mechanism of antibody-mediated blockade of TGF-ß1 activation and immunosuppression by Tregs.


Asunto(s)
Tolerancia Inmunológica , Proteínas de la Membrana/química , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/química , Humanos , Activación de Linfocitos , Proteínas de la Membrana/inmunología , Conformación Proteica en Lámina beta , Pliegue de Proteína , Factor de Crecimiento Transformador beta1/inmunología
9.
Methods Mol Biol ; 1827: 129-144, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30196495

RESUMEN

Variable fragments of heavy-chain-only antibodies (VHH) found in camelids are valuable research tools in pharmacology and biotechnology and are being developed for the clinic to treat patients with autoimmune and infectious diseases or cancer. Their single-domain nature and biochemical properties greatly facilitate the development process. The most common technology to select single-domain antibody fragments is phage display following active immunization of llamas or other members of Camelidae family. Selection of VHH from immune phage libraries is a rapid approach to discover a broad panel of in vivo matured antigen-specific clones with comprehensive functionalities. In this chapter, we describe a detailed protocol for construction of VHH immune libraries and phage display selection against antigens in their native conformation.


Asunto(s)
Técnicas de Visualización de Superficie Celular/métodos , Región Variable de Inmunoglobulina/biosíntesis , Anticuerpos de Dominio Único/biosíntesis , Animales , Bacteriófagos/genética , Secuencia de Bases , Camélidos del Nuevo Mundo , ADN Complementario/genética , Vectores Genéticos/metabolismo , Inmunización , Región Variable de Inmunoglobulina/química , Leucocitos Mononucleares/metabolismo , Reacción en Cadena de la Polimerasa , Dominios Proteicos , ARN/aislamiento & purificación , Anticuerpos de Dominio Único/química , Transformación Genética , Virión/metabolismo
10.
MAbs ; 10(4): 636-650, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29494279

RESUMEN

Identifying monoclonal antibodies that block human voltage-gated ion channels (VGICs) is a challenging endeavor exacerbated by difficulties in producing recombinant ion channel proteins in amounts that support drug discovery programs. We have developed a general strategy to address this challenge by combining high-level expression of recombinant VGICs in Tetrahymena thermophila with immunization of phylogenetically diverse species and unique screening tools that allow deep-mining for antibodies that could potentially bind functionally important regions of the protein. Using this approach, we targeted human Kv1.3, a voltage-gated potassium channel widely recognized as a therapeutic target for the treatment of a variety of T-cell mediated autoimmune diseases. Recombinant Kv1.3 was used to generate and recover 69 full-length anti-Kv1.3 mAbs from immunized chickens and llamas, of which 10 were able to inhibit Kv1.3 current. Select antibodies were shown to be potent (IC50<10 nM) and specific for Kv1.3 over related Kv1 family members, hERG and hNav1.5.


Asunto(s)
Anticuerpos Monoclonales , Descubrimiento de Drogas/métodos , Canal de Potasio Kv1.3/antagonistas & inhibidores , Animales , Camélidos del Nuevo Mundo , Pollos , Humanos , Proteínas Recombinantes , Tetrahymena thermophila
11.
J Immunol ; 199(2): 391-396, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28607112

RESUMEN

Production of active TGF-ß is regulated at a posttranslational level and implies release of the mature cytokine dimer from the inactive, latent TGF-ß precursor. There are several cell-type specific mechanisms of TGF-ß activation. We identified a new mechanism operating on the surface of human regulatory T cells and involving membrane protein GARP, which binds latent TGF-ß1. The paracrine activity of regulatory T cell-derived TGF-ß1 contributes to immunosuppression and can be inhibited with anti-GARP Abs. Whether other immune cell types use surface GARP to activate latent TGF-ß1 was not known. We show in this study that stimulated, human B lymphocytes produce active TGF-ß1 from surface GARP/latent TGF-ß1 complexes with isotype switching to IgA production.


Asunto(s)
Linfocitos B/inmunología , Inmunoglobulina A/biosíntesis , Cambio de Clase de Inmunoglobulina , Proteínas de la Membrana/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Linfocitos B/efectos de los fármacos , Citocinas/inmunología , Humanos , Inmunoglobulina A/inmunología , Activación de Linfocitos , Oligodesoxirribonucleótidos/inmunología , Unión Proteica , Recombinación Genética , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/inmunología
12.
MAbs ; 8(6): 1126-35, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27211075

RESUMEN

The identification of functional monoclonal antibodies directed against G-protein coupled receptors (GPCRs) is challenging because of the membrane-embedded topology of these molecules. Here, we report the successful combination of llama DNA immunization with scFv-phage display and selections using virus-like particles (VLP) and the recombinant extracellular domain of the GPCR glucagon receptor (GCGR), resulting in glucagon receptor-specific antagonistic antibodies. By immunizing outbred llamas with plasmid DNA containing the human GCGR gene, we sought to provoke their immune system, which generated a high IgG1 response. Phage selections on VLPs allowed the identification of mAbs against the extracellular loop regions (ECL) of GCGR, in addition to multiple VH families interacting with the extracellular domain (ECD) of GCGR. Identifying mAbs binding to the ECL regions of GCGR is challenging because the large ECD covers the small ECLs in the energetically most favorable 'closed conformation' of GCGR. Comparison of Fab with scFv-phage display demonstrated that the multivalent nature of scFv display is essential for the identification of GCGR specific clones by selections on VLPs because of avid interaction. Ten different VH families that bound 5 different epitopes on the ECD of GCGR were derived from only 2 DNA-immunized llamas. Seven VH families demonstrated interference with glucagon-mediated cAMP increase. This combination of technologies proved applicable in identifying multiple functional binders in the class B GPCR context, suggesting it is a robust approach for tackling difficult membrane proteins.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Inmunización , Epítopos Inmunodominantes/inmunología , Receptores de Glucagón/antagonistas & inhibidores , Anticuerpos de Cadena Única/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Animales , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/aislamiento & purificación , Especificidad de Anticuerpos , Péptidos Catiónicos Antimicrobianos , Células CHO , Camélidos del Nuevo Mundo/inmunología , Catelicidinas/inmunología , Técnicas de Visualización de Superficie Celular , Células Cultivadas , Cricetulus , Fibroblastos , Células HEK293 , Humanos , Fragmentos Fab de Inmunoglobulinas/sangre , Fragmentos Fab de Inmunoglobulinas/inmunología , Proteínas de la Membrana , Plásmidos/genética , Plásmidos/inmunología , Receptores de Glucagón/genética , Receptores de Glucagón/inmunología , Anticuerpos de Cadena Única/sangre
13.
Sci Transl Med ; 7(284): 284ra56, 2015 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-25904740

RESUMEN

Regulatory T cells (Tregs) are essential to prevent autoimmunity, but excessive Treg function contributes to cancer progression by inhibiting antitumor immune responses. Tregs exert contact-dependent inhibition of immune cells through the production of active transforming growth factor-ß1 (TGF-ß1). On the Treg cell surface, TGF-ß1 is in an inactive form bound to membrane protein GARP and then activated by an unknown mechanism. We demonstrate that GARP is involved in this activation mechanism. Two anti-GARP monoclonal antibodies were generated that block the production of active TGF-ß1 by human Tregs. These antibodies recognize a conformational epitope that requires amino acids GARP137-139 within GARP/TGF-ß1 complexes. A variety of antibodies recognizing other GARP epitopes did not block active TGF-ß1 production by Tregs. In a model of xenogeneic graft-versus-host disease in NSG mice, the blocking antibodies inhibited the immunosuppressive activity of human Tregs. These antibodies may serve as therapeutic tools to boost immune responses to infection or cancer via a mechanism of action distinct from that of currently available immunomodulatory antibodies. Used alone or in combination with tumor vaccines or antibodies targeting the CTLA4 or PD1/PD-L1 pathways, blocking anti-GARP antibodies may improve the efficiency of cancer immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/química , Inmunosupresores/química , Proteínas de la Membrana/química , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/química , Animales , Autoinmunidad , Epítopos/química , Enfermedad Injerto contra Huésped , Humanos , Proteínas de la Membrana/metabolismo , Metilación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Unión Proteica , Conformación Proteica , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA