Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Immunol ; 35: 285-311, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28446061

RESUMEN

IgG antibodies mediate a diversity of immune functions by coupling of antigen specificity through the Fab domain to signal transduction via Fc-Fc receptor interactions. Indeed, balanced IgG signaling through type I and type II Fc receptors is required for the control of proinflammatory, anti-inflammatory, and immunomodulatory processes. In this review, we discuss the mechanisms that govern IgG-Fc receptor interactions, highlighting the diversity of Fc receptor-mediated effector functions that regulate immunity and inflammation as well as determine susceptibility to infection and autoimmunity and responsiveness to antibody-based therapeutics and vaccines.


Asunto(s)
Anticuerpos/uso terapéutico , Enfermedades Autoinmunes/inmunología , Inmunoglobulina G/metabolismo , Inmunoterapia/métodos , Infecciones/inmunología , Receptores Fc/metabolismo , Animales , Enfermedades Autoinmunes/terapia , Susceptibilidad a Enfermedades , Humanos , Inmunidad Humoral , Infecciones/terapia , Inflamación , Transducción de Señal
2.
Cell ; 187(2): 375-389.e18, 2024 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-38242085

RESUMEN

Immune checkpoint inhibition treatment using aPD-1 monoclonal antibodies is a promising cancer immunotherapy approach. However, its effect on tumor immunity is narrow, as most patients do not respond to the treatment or suffer from recurrence. We show that the crosstalk between conventional type I dendritic cells (cDC1) and T cells is essential for an effective aPD-1-mediated anti-tumor response. Accordingly, we developed a bispecific DC-T cell engager (BiCE), a reagent that facilitates physical interactions between PD-1+ T cells and cDC1. BiCE treatment promotes the formation of active dendritic/T cell crosstalk in the tumor and tumor-draining lymph nodes. In vivo, single-cell and physical interacting cell analysis demonstrates the distinct and superior immune reprogramming of the tumors and tumor-draining lymph nodes treated with BiCE as compared to conventional aPD-1 treatment. By bridging immune cells, BiCE potentiates cell circuits and communication pathways needed for effective anti-tumor immunity.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Humanos , Anticuerpos Biespecíficos/uso terapéutico , Células Dendríticas/inmunología , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T/inmunología
3.
Nat Immunol ; 20(4): 482-492, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30833793

RESUMEN

Gut-derived antigens trigger immunoglobulin A (IgA) immune responses that are initiated by cognate B cells in Peyer's patches (PPs). These cells colonize the subepithelial domes (SEDs) of the PPs and subsequently infiltrate pre-existing germinal centers (GCs). Here we defined the pre-GC events and the micro-anatomical site at which affinity-based B cell selection occurred in PPs. Using whole-organ imaging, we showed that the affinity of the B cell antigen receptor (BCR) regulated the infiltration of antigen-specific B cells into GCs but not clonal competition in the SED. Follicular helper-like T cells resided in the SED and promoted its B cell colonization, independently of the magnitude of BCR affinity. Imaging and immunoglobulin sequencing indicated that selective clonal expansion ensued during infiltration into GCs. Thus, in contrast to the events in draining lymph nodes and spleen, in PPs, T cells promoted mainly the population expansion of B cells without clonal selection during pre-GC events. These findings have major implications for the design of oral vaccines.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Ganglios Linfáticos Agregados/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Animales , Selección Clonal Mediada por Antígenos , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Linfocitos T Colaboradores-Inductores/inmunología
4.
Nat Immunol ; 15(8): 707-16, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25045879

RESUMEN

Antibodies produced in response to a foreign antigen are characterized by polyclonality, not only in the diverse epitopes to which their variable domains bind but also in the various effector molecules to which their constant regions (Fc domains) engage. Thus, the antibody's Fc domain mediates diverse effector activities by engaging two distinct classes of Fc receptors (type I and type II) on the basis of the two dominant conformational states that the Fc domain may adopt. These conformational states are regulated by the differences among antibody subclasses in their amino acid sequence and by the complex, biantennary Fc-associated N-linked glycan. Here we discuss the diverse downstream proinflammatory, anti-inflammatory and immunomodulatory consequences of the engagement of type I and type II Fc receptors in the context of infectious, autoimmune, and neoplastic disorders.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Fragmentos Fc de Inmunoglobulinas/inmunología , Receptores de IgG/inmunología , Secuencia de Aminoácidos , Anticuerpos/inmunología , Presentación de Antígeno/inmunología , Enfermedades Autoinmunes/inmunología , Glicosilación , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/clasificación , Inmunoglobulina G/inmunología , Neoplasias/inmunología , Conformación Proteica , Estructura Terciaria de Proteína , Receptores de IgG/química , Receptores de IgG/clasificación , Vacunación
5.
Immunity ; 46(4): 577-586, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28410988

RESUMEN

CD25 is expressed at high levels on regulatory T (Treg) cells and was initially proposed as a target for cancer immunotherapy. However, anti-CD25 antibodies have displayed limited activity against established tumors. We demonstrated that CD25 expression is largely restricted to tumor-infiltrating Treg cells in mice and humans. While existing anti-CD25 antibodies were observed to deplete Treg cells in the periphery, upregulation of the inhibitory Fc gamma receptor (FcγR) IIb at the tumor site prevented intra-tumoral Treg cell depletion, which may underlie the lack of anti-tumor activity previously observed in pre-clinical models. Use of an anti-CD25 antibody with enhanced binding to activating FcγRs led to effective depletion of tumor-infiltrating Treg cells, increased effector to Treg cell ratios, and improved control of established tumors. Combination with anti-programmed cell death protein-1 antibodies promoted complete tumor rejection, demonstrating the relevance of CD25 as a therapeutic target and promising substrate for future combination approaches in immune-oncology.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Línea Celular Tumoral , Citometría de Flujo , Humanos , Inmunoterapia/métodos , Células K562 , Estimación de Kaplan-Meier , Depleción Linfocítica , Ratones , Neoplasias/patología , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Unión Proteica/inmunología , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Linfocitos T Reguladores/metabolismo
6.
Nano Lett ; 22(11): 4376-4382, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35616515

RESUMEN

Autoimmune diseases and in particular type 1 diabetes rely heavily on treatments that target the symptoms rather than prevent the underlying disease. One of the barriers to better therapeutic strategies is the inability to detect and efficiently target rare autoreactive T-cell populations that are major drivers of these conditions. Here, we develop a unique artificial antigen-presenting cell (aAPC) system from biocompatible polymer particles that allows specific encapsulation of bioactive ingredients. Using our aAPC, we demonstrate that we are able to detect rare autoreactive CD4 populations in human patients, and using mouse models, we demonstrate that our particles are able to induce desensitization in the autoreactive population. This system provides a promising tool that can be used in the prevention of autoimmunity before disease onset.


Asunto(s)
Diabetes Mellitus Tipo 1 , Linfocitos T , Animales , Células Presentadoras de Antígenos , Autoinmunidad , Linfocitos T CD4-Positivos , Diabetes Mellitus Tipo 1/terapia , Humanos , Ratones
7.
Proc Natl Acad Sci U S A ; 115(43): 11048-11053, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30297432

RESUMEN

Immune stimulation has emerged as a promising approach to the treatment of neoplastic diseases. Currently approved therapeutics, such as anti-CTLA4 and anti-PD1, are primarily aimed at blocking inhibitory signaling by immune cells. An alternative and potentially synergistic approach would involve activation of immune pathways by agonism of stimulatory receptors, such as CD40. Agonistic antibodies, while promising in principle, have encountered significant barriers in clinical trials limited by the systemic toxicity of such approaches. Using a mouse model humanized for both Fc receptors and CD40, we previously demonstrated enhanced antitumor activity with an Fc-modified antibody. We now demonstrate that this model recapitulates the platelet and hepatic toxicities seen with anti-CD40 antibodies in patients, providing a predictive measure of the dose-limiting activity of this approach. We further show that such toxicity can be circumvented and durable systemic antitumor immunity achieved by intratumoral delivery of an Fc-engineered anti-CD40 agonistic antibody.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos CD40/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Animales , Antígeno CTLA-4/inmunología , Línea Celular Tumoral , Humanos , Inmunoterapia/métodos , Inyecciones Intralesiones/métodos , Ratones
8.
J Immunol ; 192(9): 4164-73, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24683185

RESUMEN

CD74, the cell-surface form of the MHC class II invariant chain, is a key inflammatory factor that is involved in various immune-mediated diseases as part of the macrophage migration inhibitory factor (MIF) binding complex. However, little is known about the natural regulators of CD74 in this context. In order to study the role of the HLA-DR molecule in regulating CD74, we used the HLA-DRα1 domain, which was shown to bind to and downregulate CD74 on CD11b(+) monocytes. We found that DRα1 directly inhibited binding of MIF to CD74 and blocked its downstream inflammatory effects in the spinal cord of mice with experimental autoimmune encephalomyelitis (EAE). Potency of the DRα1 domain could be destroyed by trypsin digestion but enhanced by addition of a peptide extension (myelin oligodendrocyte glycoprotein [MOG]-35-55 peptide) that provided secondary structure not present in DRα1. These data suggest a conformationally sensitive determinant on DRα1-MOG that is responsible for optimal binding to CD74 and antagonism of MIF effects, resulting in reduced axonal damage and reversal of ongoing clinical and histological signs of EAE. These results demonstrate natural antagonist activity of DRα1 for MIF that was strongly potentiated by the MOG peptide extension, resulting in a novel therapeutic, DRα1-MOG-35-55, that within the limitations of the EAE model may have the potential to treat autoimmune diseases such as multiple sclerosis.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Cadenas alfa de HLA-DR/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Animales , Western Blotting , Citometría de Flujo , Humanos , Ratones , Ratones Transgénicos , Monocitos/metabolismo , Médula Espinal/metabolismo
9.
Cancer Immunol Res ; 12(3): 322-333, 2024 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-38147316

RESUMEN

Preclinical murine data indicate that fragment crystallizable (Fc)-dependent depletion of intratumoral regulatory T cells (Treg) is a major mechanism of action of anti-CTLA-4. However, the two main antibodies administered to patients (ipilimumab and tremelimumab) do not recapitulate these effects. Here, we investigate the underlying mechanisms responsible for the limited Treg depletion observed with these therapies. Using an immunocompetent murine model humanized for CTLA-4 and Fcγ receptors (FcγR), we show that ipilimumab and tremelimumab exhibit limited Treg depletion in tumors. Immune profiling of the tumor microenvironment (TME) in both humanized mice and humans revealed high expression of the inhibitory Fc receptor, FcγRIIB, which limits antibody-dependent cellular cytotoxicity/phagocytosis. Blocking FcγRIIB in humanized mice rescued the Treg-depleting capacity and antitumor activity of ipilimumab. Furthermore, Fc engineering of antibodies targeting Treg-associated targets (CTLA-4 or CCR8) to minimize FcγRIIB binding significantly enhanced Treg depletion, resulting in increased antitumor activity across various tumor models. Our results define the inhibitory FcγRIIB as an immune checkpoint limiting antibody-mediated Treg depletion in the TME, and demonstrate Fc engineering as an effective strategy to overcome this limitation and improve the efficacy of Treg-targeting antibodies.


Asunto(s)
Neoplasias , Linfocitos T Reguladores , Humanos , Animales , Ratones , Ipilimumab/farmacología , Ipilimumab/uso terapéutico , Antígeno CTLA-4 , Microambiente Tumoral , Neoplasias/tratamiento farmacológico
10.
Nat Cancer ; 5(5): 742-759, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38429414

RESUMEN

Successful immunotherapy relies on triggering complex responses involving T cell dynamics in tumors and the periphery. Characterizing these responses remains challenging using static human single-cell atlases or mouse models. To address this, we developed a framework for in vivo tracking of tumor-specific CD8+ T cells over time and at single-cell resolution. Our tools facilitate the modeling of gene program dynamics in the tumor microenvironment (TME) and the tumor-draining lymph node (tdLN). Using this approach, we characterize two modes of anti-programmed cell death protein 1 (PD-1) activity, decoupling induced differentiation of tumor-specific activated precursor cells from conventional type 1 dendritic cell (cDC1)-dependent proliferation and recruitment to the TME. We demonstrate that combining anti-PD-1 therapy with anti-4-1BB agonist enhances the recruitment and proliferation of activated precursors, resulting in tumor control. These data suggest that effective response to anti-PD-1 therapy is dependent on sufficient influx of activated precursor CD8+ cells to the TME and highlight the importance of understanding system-level dynamics in optimizing immunotherapies.


Asunto(s)
Linfocitos T CD8-positivos , Inmunoterapia , Microambiente Tumoral , Animales , Ratones , Inmunoterapia/métodos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Microambiente Tumoral/inmunología , Humanos , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Células Dendríticas/inmunología , Células Dendríticas/efectos de los fármacos , Línea Celular Tumoral
11.
J Autoimmun ; 47: 83-93, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24090977

RESUMEN

The trimolecular complex composed of autoreactive T-cell receptor, MHC class II, and an autoantigenic peptide plays a central role in the activation of pathogenic Islet-specific CD4+ T cells in type 1 diabetes (T1D). We isolated and characterized novel antibodies against autoreactive T-cell epitopes associated with T1D. Our antibodies mimic the specificity of the T-cell receptor (TCR), while binding MHC class II/peptide complexes in an autoantigen peptide specific, MHC-restricted manner. The isolated TCR-like antibodies were directed against the minimal T-cell epitope GAD-555-567 in the context of the HLA-DR4-diabetic-associated molecule. A representative high-affinity TCR-like antibody clone (G3H8) enabled the detection of intra- and extra-cellular DR4/GAD-555-567 complexes in antigen presenting cells. I561M single mutation at the central position (P5) of the GAD-555-567 peptide abolished the binding of G3H8 to the DR4/GAD complex, demonstrating its high fine TCR-like specificity. The G3H8 TCR-like antibody significantly inhibited GAD-555-567 specific, DR4 restricted T-cell response in vitro and in vivo in HLA-DR4 transgenic mice. Our findings constitute a proof-of-concept for the utility of TCR-like antibodies as antigen-specific immunomodulation agents for regulating pathogenic T-cells and suggest that TCR-like antibodies targeting autoreactive MHC class II epitopes are valuable research tools that enable studies related to antigen presentation as well as novel therapeutic agents that may be used to modulate autoimmune disorders such as T1D.


Asunto(s)
Anticuerpos/uso terapéutico , Diabetes Mellitus Tipo 1/terapia , Epítopos de Linfocito T/inmunología , Glutamato Descarboxilasa/inmunología , Inmunomodulación , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes/uso terapéutico , Animales , Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/inmunología , Autoantígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Línea Celular , Proliferación Celular , Epítopos de Linfocito T/genética , Glutamato Descarboxilasa/genética , Células HEK293 , Antígeno HLA-DR4/genética , Humanos , Inmunoglobulina G/inmunología , Interleucina-2/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Unión Proteica/genética , Unión Proteica/inmunología
12.
J Autoimmun ; 40: 96-110, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23026773

RESUMEN

Treatment with partial (p)MHC class II-ß1α1 constructs (also referred to as recombinant T-cell receptor ligands - RTL) linked to antigenic peptides can induce T-cell tolerance, inhibit recruitment of inflammatory cells and reverse autoimmune diseases. Here we demonstrate a novel regulatory pathway that involves RTL binding to CD11b(+) mononuclear cells through a receptor comprised of MHC class II invariant chain (CD74), cell-surface histones and MHC class II itself for treatment of experimental autoimmune encephalomyelitis (EAE). Binding of RTL constructs with CD74 involved a previously unrecognized MHC class II-α1/CD74 interaction that inhibited CD74 expression, blocked activity of its ligand, macrophage migration inhibitory factor, and reduced EAE severity. These findings implicate binding of RTL constructs to CD74 as a key step in both antigen-driven and bystander T-cell tolerance important in treatment of inflammatory diseases.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/metabolismo , Enfermedades Autoinmunes/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Monocitos/inmunología , Animales , Antígenos de Diferenciación de Linfocitos B/biosíntesis , Antígeno CD11b/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Antígenos de Histocompatibilidad Clase II/biosíntesis , Antígenos de Histocompatibilidad Clase II/inmunología , Tolerancia Inmunológica , Oxidorreductasas Intramoleculares , Factores Inhibidores de la Migración de Macrófagos , Proteínas de la Membrana , Ratones , Receptores de Antígenos de Linfocitos T , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/inmunología
13.
Sci Immunol ; 8(81): eadd8005, 2023 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-36867679

RESUMEN

FDA-approved anti-PD-L1 monoclonal antibodies (mAbs) bear the IgG1 isotype, whose scaffolds are either wild-type (e.g., avelumab) or Fc-mutated and lacking Fcγ receptor (FcγR) engagement (e.g., atezolizumab). It is unknown whether variation in the ability of the IgG1 Fc region to engage FcγRs renders mAbs with superior therapeutic activity. In this study, we used humanized FcγR mice to study the contribution of FcγR signaling to the antitumor activity of human anti-PD-L1 mAbs and to identify an optimal human IgG scaffold for PD-L1 mAbs. We observed similar antitumor efficacy and comparable tumor immune responses in mice treated with anti-PD-L1 mAbs with wild-type and Fc-mutated IgG scaffolds. However, in vivo antitumor activity of the wild-type anti-PD-L1 mAb avelumab was enhanced by combination treatment with an FcγRIIB-blocking antibody, which was co-administered to overcome the suppressor function of FcγRIIB in the tumor microenvironment (TME). We performed Fc glycoengineering to remove the fucose subunit from the Fc-attached glycan of avelumab to enhance its binding to the activating FcγRIIIA. Treatment with the Fc-afucosylated version of avelumab also enhanced antitumor activity and induced stronger antitumor immune responses compared with the parental IgG. The enhanced effect by afucosylated PD-L1 antibody was dependent on neutrophils and associated with decreased frequencies of PD-L1+ myeloid cells and increased infiltration of T cells in the TME. Our data reveal that the current design of FDA-approved anti-PD-L1 mAbs does not optimally harness FcγR pathways and suggest two strategies to enhance FcγR engagement to optimize anti-PD-L1 immunotherapy.


Asunto(s)
Anticuerpos Monoclonales , Receptores de IgG , Humanos , Animales , Ratones , Inmunoglobulina G , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia
14.
bioRxiv ; 2023 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-36711504

RESUMEN

Despite pre-clinical murine data supporting T regulatory (Treg) cell depletion as a major mechanism by which anti-CTLA-4 antibodies function in vivo, the two main antibodies tested in patients (ipilimumab and tremelimumab) have failed to demonstrate similar effects. We report analogous findings in an immunocompetent murine model humanized for CTLA-4 and Fcy receptors (hCTLA-4/hFcyR mice), where both ipilimumab and tremelimumab fail to show appreciable Treg depletion. Immune profiling of the tumor microenvironment (TME) in both mice and human samples revealed upregulation of the inhibitory Fcy receptor, FcyRIIB, which limits the ability of the antibody Fc fragment of human anti-CTLA-4 antibodies to induce effective antibody dependent cellular cytotoxicty/phagocytosis (ADCC/ADCP). Blocking FcyRIIB in humanized mice rescues Treg depleting capacity and anti-tumor activity of ipilimumab. For another target, CC motif chemokine receptor 8 (CCR8), which is selectively expressed on tumor infiltrating Tregs, we show that Fc engineering to enhance binding to activating Fc receptors, while limiting binding to the inhibitory Fc receptor, leads to consistent Treg depletion and single-agent activity across multiple tumor models, including B16, MC38 and MB49. These data reveal the importance of reducing engagement to the inhibitory Fc receptor to optimize Treg depletion by TME targeting antibodies. Our results define the inhibitory FcyRIIB receptor as a novel immune checkpoint limiting antibody-mediated Treg depletion in tumors, and demonstrate Fc variant engineering as a means to overcome this limitation and augment efficacy for a repertoire of antibodies currently in use or under clinical evaluation in oncology.

15.
Eur J Immunol ; 41(5): 1465-79, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21469129

RESUMEN

Antigen-presenting cell-associated four-domain MHC class II (MHC-II) molecules play a central role in activating autoreactive CD4(+) T cells involved in multiple sclerosis (MS) and type 1 diabetes (T1D). In contrast, two-domain MHC-II structures with the same covalently attached self-peptide (recombinant T-cell receptor ligands (RTLs)) can regulate pathogenic CD4(+) T cells and reverse clinical signs of experimental autoimmune diseases. RTL1000, which is composed of the ß1α1 domains of human leukocyte antigen (HLA)-DR2 linked to the encephalitogenic human myelin oligodendrocyte glycoprotein (MOG)-35-55 peptide, was recently shown to be safe and well tolerated in a phase I clinical trial in MS. To evaluate the opposing biological effects of four- versus two-domain MHC-II structures, we screened phage Fab antibodies (Abs) for the neutralizing activity of RTL1000. Five different TCR-like Abs were identified that could distinguish between the two- versus four-domain MHC-peptide complexes while the cognate TCR was unable to make such a distinction. Moreover, Fab detection of native two-domain HLA-DR structures in human plasma implies that there are naturally occurring regulatory MHC-peptide complexes. These results demonstrate for the first time distinct conformational determinants characteristic of activating versus tolerogenic MHC-peptide complexes involved in human autoimmunity.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Autoinmunidad/inmunología , Diabetes Mellitus Tipo 1/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Glicoproteínas , Antígeno HLA-DR2/química , Antígeno HLA-DR2/genética , Antígeno HLA-DR2/inmunología , Humanos , Tolerancia Inmunológica , Fragmentos Fab de Inmunoglobulinas/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Ratones , Ratones Transgénicos , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/terapia , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos , Proteínas Recombinantes de Fusión/sangre , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes/inmunología
16.
Expert Rev Mol Med ; 14: e6, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22361332

RESUMEN

Tumour and virus-infected cells are recognised by CD8+ cytotoxic T cells that, in response, are activated to eliminate these cells. In order to be activated, the clonotypic T-cell receptor (TCR) needs to encounter a specific peptide antigen presented by the membrane surface major histocompatibility complex (MHC) molecule. Cells that have undergone malignant transformation or viral infection present peptides derived from tumour-associated antigens or viral proteins on their MHC class I molecules. Therefore, disease-specific MHC-peptide complexes are desirable targets for immunotherapeutic approaches. One such approach transforms the unique fine specificity but low intrinsic affinity of TCRs to MHC-peptide complexes into high-affinity soluble antibody molecules endowed with a TCR-like specificity towards tumour or viral epitopes. These antibodies, termed TCR-like antibodies, are being developed as a new class of immunotherapeutics that can target tumour and virus-infected cells and mediate their specific killing. In addition to their therapeutic capabilities, TCR-like antibodies are being developed as diagnostic reagents for cancer and infectious diseases, and serve as valuable research tools for studying MHC class I antigen presentation.


Asunto(s)
Antígenos de Neoplasias/uso terapéutico , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Receptores de Antígenos de Linfocitos T , Virosis/tratamiento farmacológico , Animales , Afinidad de Anticuerpos/inmunología , Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/patología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Proteínas de Neoplasias/inmunología , Neoplasias/inmunología , Neoplasias/patología , Péptidos/inmunología , Proteínas Virales/inmunología , Virosis/inmunología , Virosis/patología
17.
Front Immunol ; 13: 940674, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35911742

RESUMEN

The clinical use of anti-CD40 agonist monoclonal antibodies (mAbs) is aimed at recruiting the immune system to fight the tumor cells. This approach has been demonstrated to be effective in various preclinical models. However, human CD40 Abs displayed only modest antitumor activity in cancer patients, characterized by low efficacy and dose-limiting toxicity. While recent studies highlight the importance of engineering the Fc region of human CD40 mAbs to optimize their agonistic potency, toxicity remains the main limiting factor, restricting clinical application to suboptimal doses. Here, we discuss the current challenges in realizing the full potential of CD40 mAbs in clinical practice, and describe novel approaches designed to circumvent the systemic toxicity associated with CD40 agonism.


Asunto(s)
Antineoplásicos Inmunológicos , Antineoplásicos , Antígenos CD40 , Neoplasias , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Antígenos CD40/inmunología , Línea Celular Tumoral , Humanos , Inmunoterapia , Neoplasias/terapia
18.
Nat Cancer ; 3(3): 287-302, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35190724

RESUMEN

Therapeutic use of agonistic anti-CD40 antibodies is a potentially powerful approach for activation of the immune response to eradicate tumors. However, the translation of this approach to clinical practice has been substantially restricted due to the severe dose-limiting toxicities observed in multiple clinical trials. Here, we demonstrate that conventional type 1 dendritic cells are essential for triggering antitumor immunity but not the toxicity of CD40 agonists, while macrophages, platelets and monocytes lead to toxic events. Therefore, we designed bispecific antibodies that target CD40 activation preferentially to dendritic cells, by coupling the CD40 agonist arm with CD11c-, DEC-205- or CLEC9A-targeting arms. These bispecific reagents demonstrate a superior safety profile compared to their parental CD40 monospecific antibody while triggering potent antitumor activity. We suggest such cell-selective bispecific agonistic antibodies as a drug platform to bypass the dose-limiting toxicities of anti-CD40, and of additional types of agonistic antibodies used for cancer immunotherapy.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Anticuerpos Biespecíficos/farmacología , Antígenos CD40 , Células Dendríticas , Humanos , Inmunoterapia , Neoplasias/tratamiento farmacológico
19.
Sci Adv ; 8(8): eabm4552, 2022 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-35213218

RESUMEN

GITR is a TNF receptor, and its activation promotes immune responses and drives antitumor activity. The receptor is activated by the GITR ligand (GITRL), which is believed to cluster receptors into a high-order array. Immunotherapeutic agonist antibodies also activate the receptor, but their mechanisms are not well characterized. We solved the structure of full-length mouse GITR bound to Fabs from the antibody DTA-1. The receptor is a dimer, and each subunit binds one Fab in an orientation suggesting that the antibody clusters receptors. Binding experiments with purified proteins show that DTA-1 IgG and GITRL both drive extensive clustering of GITR. Functional data reveal that DTA-1 and the anti-human GITR antibody TRX518 activate GITR in their IgG forms but not as Fabs. Thus, the divalent character of the IgG agonists confers an ability to mimic GITRL and cluster and activate GITR. These findings will inform the clinical development of this class of antibodies for immuno-oncology.

20.
Cell Rep ; 37(11): 110114, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34883043

RESUMEN

Messenger RNA-based vaccines against COVID-19 induce a robust anti-SARS-CoV-2 antibody response with potent viral neutralization activity. Antibody effector functions are determined by their constant region subclasses and by their glycosylation patterns, but their role in vaccine efficacy is unclear. Moreover, whether vaccination induces antibodies similar to those in patients with COVID-19 remains unknown. We analyze BNT162b2 vaccine-induced IgG subclass distribution and Fc glycosylation patterns and their potential to drive effector function via Fcγ receptors and complement pathways. We identify unique and dynamic pro-inflammatory Fc compositions that are distinct from those in patients with COVID-19 and convalescents. Vaccine-induced anti-Spike IgG is characterized by distinct Fab- and Fc-mediated functions between different age groups and in comparison to antibodies generated during natural viral infection. These data highlight the heterogeneity of Fc responses to SARS-CoV-2 infection and vaccination and suggest that they support long-lasting protection differently.


Asunto(s)
COVID-19/inmunología , Glicosilación/efectos de los fármacos , SARS-CoV-2/inmunología , Adulto , Anciano , Anticuerpos Antivirales/inmunología , Vacuna BNT162/inmunología , Vacunas contra la COVID-19/metabolismo , Femenino , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Israel/epidemiología , Masculino , Persona de Mediana Edad , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunación/métodos , Eficacia de las Vacunas , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/metabolismo , Vacunas de ARNm/inmunología , Vacunas de ARNm/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA