Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Immunother Cancer ; 3: 37, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26500773

RESUMEN

BACKGROUND: Clinical targeting of TNFR family of receptors (CD40, CD134 and CD137) with immunostimulatory monoclonal antibodies has been successful in cancer immunotherapy. However, targeting of CD27 with a mAb is a relatively new approach to provide costimulation of immune cells undergoing activation. Thus, activation of human CD27 (TNFRSF7) with a monoclonal antibody (varlilumab) has previously been demonstrated to result in T cell activation and anti-tumor activity in preclinical models, and is currently in early phase clinical trials in patients with advanced malignancies. In this study we used an in vitro system using human peripheral blood T cells to characterize the varlilumab-mediated costimulatory effects in combination with TCR stimulation in terms of phenotypic, transcriptional and functionality changes. METHODS: T cells were isolated from normal volunteer PBMCs using magnetic bead isolation kits and stimulated in vitro with plate bound anti-CD3 Ab (OKT3) and varlilumab or control Ab for 72 h. Activation profiles were monitored by ELISA or Luminex-based testing cytokine/chemokine releases, cell surface phenotyping for costimulatory and coinhibitory markers and CFSE dye dilution by proliferating T cells and Tregs. Changes in gene expression and transcriptome analysis of varlilumab-stimulated T cells was carried on Agilent Human whole genome microarray datasets using a suite of statistical and bioinformatic software tools. RESULTS: Costimulation of T cells with varlilumab required continuous TCR signaling as pre-activated T cells were unable to produce cytokines with CD27 signaling alone. Analysis of T cell subsets further revealed that memory CD4+ and CD8+ T cells were specifically activated with a bias toward CD8+ T lymphocyte proliferation. Activation was accompanied by upregulated cell surface expression of costimulatory [4-1BB, OX40, GITR and ICOS] and coinhibitory [PD-1] molecules. Importantly, varlilumab costimulation did not activate purified Tregs as measured by cytokine production, proliferation and suppression of dividing non-Treg T cells. Analysis of changes in gene expression during varlilumab stimulation of T cells revealed modulation of pro-inflammatory signatures consistent with cellular activation and proliferation, with the IL-2 pathway showing the highest frequency of gene modulation. CONCLUSIONS: Altogether, the data reveal the requirements and T cell subtype-specific effects of CD27 costimulation, and helps select relevant biomarkers for studying the effects of varlilumab in patients.

2.
Sci Transl Med ; 6(232): 232ra51, 2014 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-24739759

RESUMEN

Immune-based therapies for cancer are generating substantial interest because of the success of immune checkpoint inhibitors. This study aimed to enhance anticancer immunity by exploiting the capacity of dendritic cells (DCs) to initiate T cell immunity by efficient uptake and presentation of endocytosed material. Delivery of tumor-associated antigens to DCs using receptor-specific monoclonal antibodies (mAbs) in the presence of DC-activating agents elicits robust antigen-specific immune responses in preclinical models. DEC-205 (CD205), a molecule expressed on DCs, has been extensively studied for its role in antigen processing and presentation. CDX-1401 is a vaccine composed of a human mAb specific for DEC-205 fused to the full-length tumor antigen NY-ESO-1. This phase 1 trial assessed the safety, immunogenicity, and clinical activity of escalating doses of CDX-1401 with the Toll-like receptor (TLR) agonists resiquimod (TLR7/8) and Hiltonol (poly-ICLC, TLR3) in 45 patients with advanced malignancies refractory to available therapies. Treatment induced humoral and cellular immunity to NY-ESO-1 in patients with confirmed NY-ESO-1-expressing tumors across various dose levels and adjuvant combinations. No dose-limiting or grade 3 toxicities were reported. Thirteen patients experienced stabilization of disease, with a median duration of 6.7 months (range, 2.4+ to 13.4 months). Two patients had tumor regression (~20% shrinkage in target lesions). Six of eight patients who received immune-checkpoint inhibitors within 3 months after CDX-1401 administration had objective tumor regression. This first-in-human study of a protein vaccine targeting DCs demonstrates its feasibility, safety, and biological activity and provides rationale for combination immunotherapy strategies including immune checkpoint blockade.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Epítopos/inmunología , Inmunidad Humoral/inmunología , Lectinas Tipo C/inmunología , Proteínas de la Membrana/inmunología , Receptores de Superficie Celular/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/farmacocinética , Citocinas/metabolismo , Relación Dosis-Respuesta Inmunológica , Femenino , Humanos , Inmunidad Celular/inmunología , Inmunoglobulina G/inmunología , Interferón gamma/biosíntesis , Subgrupos Linfocitarios/metabolismo , Masculino , Persona de Mediana Edad , Antígenos de Histocompatibilidad Menor , Linfocitos T/inmunología , Vacunación
3.
Clin Cancer Res ; 17(14): 4844-53, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21632857

RESUMEN

PURPOSE: The use of tumor-derived proteins as cancer vaccines is complicated by tolerance to these self-antigens. Tolerance may be broken by immunization with activated, autologous, ex vivo generated and antigen-loaded, antigen-presenting cells (APC); however, targeting tumor antigen directly to APC in vivo would be a less complicated strategy. We wished to test whether targeted delivery of an otherwise poorly immunogenic, soluble antigen to APC through their mannose receptors (MR) would induce clinically relevant immunity. EXPERIMENTAL DESIGN: Two phase I studies were conducted with CDX-1307, a vaccine composed of human chorionic gonadotropin beta-chain (hCG-ß) fused to an MR-specific monoclonal antibody, administered either locally (intradermally) or systemically (intravenously) in patients with advanced epithelial malignancies. An initial dose escalation of single-agent CDX-1307 was followed by additional cohorts of CDX-1307 combined with granulocyte-macrophage colony-stimulating factor (GM-CSF) and the Toll-like receptor (TLR) 3 agonist polyinosinic-polycytidylic acid (poly-ICLC) and TLR7/8 agonist resiquimod to activate the APC. RESULTS: CDX-1307 induced consistent humoral and T-cell responses to hCG-ß when coadministered with TLR agonists. Greater immune responses and clinical benefit, including the longest duration of stable disease, were observed with immunization combined with local TLR agonists. Immune responses were induced equally efficiently in patients with elevated and nonelevated levels of serum hCG-ß. Antibodies within the serum of vaccinated participants had tumor suppressive function in vitro. Toxicity consisted chiefly of mild injection site reactions. CONCLUSIONS: APC targeting and activation induce adaptive immunity against poorly immunogenic self-antigens which has implications for enhancing the efficacy of cancer immunotherapy.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Autoantígenos/inmunología , Vacunas contra el Cáncer/uso terapéutico , Neoplasias/inmunología , Neoplasias/terapia , Proteínas Recombinantes de Fusión/uso terapéutico , Receptores Toll-Like/agonistas , Células Presentadoras de Antígenos/metabolismo , Autoantígenos/metabolismo , Vacunas contra el Cáncer/farmacocinética , Vacunas contra el Cáncer/toxicidad , Línea Celular Tumoral , Gonadotropina Coriónica Humana de Subunidad beta/sangre , Gonadotropina Coriónica Humana de Subunidad beta/inmunología , Femenino , Humanos , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Masculino , Estadificación de Neoplasias , Neoplasias/patología , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/toxicidad , Piel/inmunología , Piel/metabolismo , Piel/patología , Receptores Toll-Like/metabolismo , Resultado del Tratamiento
4.
J Immunol ; 186(2): 1218-27, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21149605

RESUMEN

Immunization of cancer patients with vaccines containing full-length tumor Ags aims to elicit specific Abs and both CD4(+) and CD8(+) T cells. Vaccination with protein Ags, however, often elicits only CD4(+) T cell responses without inducing Ag-specific CD8(+) T cells, as exogenous protein is primarily presented to CD4(+) T cells. Recent data revealed that Ab-mediated targeting of protein Ags to cell surface receptors on dendritic cells could enhance the induction of both CD4(+) and CD8(+) T cells. We investigated in this study if these observations were applicable to NY-ESO-1, a cancer-testis Ag widely used in clinical cancer vaccine trials. We generated two novel targeting proteins consisting of the full-length NY-ESO-1 fused to the C terminus of two human mAbs against the human mannose receptor and DEC-205, both internalizing molecules expressed on APC. These targeting proteins were evaluated for their ability to activate NY-ESO-1-specific human CD4(+) and CD8(+) T cells in vitro. Both targeted NY-ESO-1 proteins rapidly bound to their respective targets on APC. Whereas nontargeted and Ab-targeted NY-ESO-1 proteins similarly activated CD4(+) T cells, cross-presentation to CD8(+) T cells was only efficiently induced by targeted NY-ESO-1. In addition, both mannose receptor and DEC-205 targeting elicited specific CD4(+) and CD8(+) T cells from PBLs of cancer patients. Receptor-specific delivery of NY-ESO-1 to APC appears to be a promising vaccination strategy to efficiently generate integrated and broad Ag-specific immune responses against NY-ESO-1 in cancer patients.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Lectinas Tipo C/metabolismo , Lectinas de Unión a Manosa/metabolismo , Proteínas de la Membrana/inmunología , Receptores de Superficie Celular/metabolismo , Secuencia de Aminoácidos , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Línea Celular , Células Clonales , Reactividad Cruzada/genética , Reactividad Cruzada/inmunología , Pruebas Inmunológicas de Citotoxicidad , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Receptor de Manosa , Lectinas de Unión a Manosa/genética , Lectinas de Unión a Manosa/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Antígenos de Histocompatibilidad Menor , Datos de Secuencia Molecular , Unión Proteica/inmunología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo
5.
Vaccine ; 26(25): 3092-102, 2008 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-18353510

RESUMEN

Dendritic cells (DCs) are highly specialized antigen-presenting cells of the immune system that are efficient at presenting peptide-antigen for the activation of T cells and are often the cell type of choice for vaccine targeting by virtue of high expression levels of MHC and costimulatory molecules. Since the level of peptide-MHC complex significantly influences stimulation of T cells, a proof-of-concept potency assay was developed to directly examine the presentation and density of MHC class I peptides derived from the processing of a model tumor antigen, (hCGbeta), on the surface of DCs. In this study we first generated antibodies (TCR mimics or TCRm) to two peptide-HLA-A*0201 epitopes derived from hCGbeta designated as TMT (40-48) and GVL (47-55). Characterization of each TCRm by ELISA and flow cytometric analysis, demonstrated specific binding to soluble recombinant HLA-A2 protein and HLA-A2.1+ T2 cells loaded with relevant peptide. TCRm reactive against the TMT and GVL epitopes blocked granzyme-B production by peptide-specific cytotoxic T lymphocytes (CTL) lines further supporting their recognition specificity. For the assessment of antigen presentation function, human immature monocyte-derived DCs (iDCs) were treated with the mannose receptor targeting vaccine, B11-hCGbeta and matured with Poly I:C. The TMT and GVL epitope reactive CTL lines responded to vaccine-treated but not vehicle-treated mature DCs (mDCs) with TMT and GVL TCRm specifically blocking IFN-gamma production. The TCRm were then used to directly confirm specific peptide-MHC complexes on mDCs. TCRm staining of vaccine-treated mDCs showed detection of the TMT and GVL peptide-HLA-A2 complexes. These findings demonstrate that TCRms may be important tools for determining the potency of DC-based vaccines.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/administración & dosificación , Epítopos/inmunología , Antígeno HLA-A2/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Vacunas contra el Cáncer/inmunología , Línea Celular , Células Dendríticas/inmunología , Epítopos/metabolismo , Antígeno HLA-A2/genética , Humanos
6.
J Transl Med ; 5: 5, 2007 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-17254349

RESUMEN

Previously, we have successfully targeted the mannose receptor (MR) expressed on monocyte-derived dendritic cells (DCs) using a fully human MR-specific antibody, B11, as a vehicle to deliver whole protein tumor antigens such as the human chorionic gonadotropin hormone (hCGbeta). Since MRs play a role in bridging innate immunity with adaptive immunity we have explored several toll-like receptor (TLR)-specific ligands that may synergize with MR targeting and be applicable as adjuvants in the clinic. We demonstrate that antigen-specific helper and cytolytic T cells from both healthy donors and cancer patients were effectively primed with B11-hCGbeta-treated autologous DCs when a combination of one or several TLR ligands is used. Specifically, concomitant signaling of DCs via TLR3 with dsRNA (poly I:C) and DC TLR 7/8 with Resiquimod (R-848), respectively, elicited efficient antigen presentation-mediated by MR-targeting. We demonstrate that MR and TLRs contribute towards maturation and activation of DCs by a mechanism that may be driven by a combination of adjuvant and antibody vaccines that specifically deliver antigenic targets to DCs.


Asunto(s)
Anticuerpos/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Inmunidad Celular/inmunología , Receptores Toll-Like/inmunología , Antígenos de Neoplasias/inmunología , Compartimento Celular/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/inmunología , Gonadotropina Coriónica Humana de Subunidad beta/inmunología , Citocinas/biosíntesis , Células Dendríticas/efectos de los fármacos , Epítopos/inmunología , Humanos , Imidazoles/farmacología , Inmunidad Celular/efectos de los fármacos , Lectinas Tipo C/inmunología , Ligandos , Receptor de Manosa , Lectinas de Unión a Manosa/inmunología , Fenotipo , Poli I-C/farmacología , Receptores de Superficie Celular/inmunología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Células TH1/inmunología
7.
Expert Opin Biol Ther ; 4(12): 1953-62, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15571457

RESUMEN

Targeting antigens to endocytic receptors on professional antigen-presenting cells (APCs) represents an attractive strategy to enhance the efficacy of vaccines. Such APC-targeted vaccines have an exceptional ability to guide exogenous protein antigens into vesicles that efficiently process the antigen for major histocompatibility complex class I and class II presentation. Efficient targeting not only requires high specificity for the receptor that is abundantly expressed on the surface of APCs, but also the ability to be rapidly internalised and loaded into compartments that contain elements of the antigen-processing machinery. The mannose receptor (MR) and related C-type lectin receptors are particularly designed to sample antigens (self and non-self), much like pattern recognition receptors, to integrate the innate with adaptive immune responses. In fact, a variety of approaches involving delivery of antigens to the MR have demonstrated effective induction of potent cellular and humoral immune responses. Yet, although several lines of evidence in diverse experimental systems attest to the efficacy of targeted vaccine strategies, it is becoming increasingly clear that additional signals, such as those afforded by adjuvants, may be critical to elicit sustained immunity. Therefore, MR-targeted vaccines are likely to be most efficacious in vivo when combined with agents that elicit complementary activation signals. Certainly, a better understanding of the mechanism associated with the induction of immune responses as a result of targeting antigens to the MR, will be important in exploiting MR-targeted vaccines not only for mounting immune defenses against cancer and infectious disease, but also for specific induction of tolerance in the treatment of autoimmune disease.


Asunto(s)
Lectinas Tipo C/metabolismo , Lectinas de Unión a Manosa/metabolismo , Receptores de Superficie Celular/metabolismo , Tecnología Farmacéutica/métodos , Vacunas/metabolismo , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/prevención & control , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Lectinas Tipo C/inmunología , Receptor de Manosa , Lectinas de Unión a Manosa/inmunología , Receptores de Superficie Celular/inmunología , Vacunas/inmunología , Vacunas/uso terapéutico
8.
Clin Cancer Res ; 10(6): 1920-7, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15041707

RESUMEN

PURPOSE: The oncofetal antigen, human chorionic gonadotropin beta subunit (hCGbeta), is expressed by a number of carcinomas and is a prognostic indicator in renal, colorectal, bladder, and pancreatic cancers. We describe the development of a novel antibody-based dendritic cell (DC)-targeted cancer vaccine capable of eliciting cellular immune responses directed against hCGbeta. EXPERIMENTAL DESIGN: The tumor-associated antigen hCGbeta was coupled genetically to a human anti-DC antibody (B11). The resulting fusion protein (B11-hCGbeta) was evaluated for its ability to promote tumor antigen-specific cellular immune responses in a human in vitro model. Monocyte-derived human DCs from normal donors were exposed to purified B11-hCGbeta, activated with CD40 ligand, mixed with autologous lymphocytes, and tested for their ability to promote hCGbeta-specific proliferative and cytotoxic T-lymphocyte responses. RESULTS: B11-hCGbeta was found to be a soluble, well-defined, and readily purified product that specifically recognized the human mannose receptor via the B11 antibody portion of the fusion protein. B11-hCGbeta functionally promoted the uptake and processing of tumor antigen by DCs, which led to the generation of tumor-specific HLA class I and class II-restricted T-cell responses, including CTLs capable of killing human cancer cell lines expressing hCGbeta. CONCLUSIONS: Although other hCG vaccines have been shown to be capable of eliciting antibody responses to hCGbeta, this is the first time that cellular immune responses to hCGbeta have been induced by a vaccine in a human system. This DC-targeted hCGbeta vaccine holds promise for the management of a number of cancers and merits additional clinical development.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/farmacología , Gonadotropina Coriónica Humana de Subunidad beta/inmunología , Células Dendríticas/inmunología , Animales , Antígenos de Neoplasias/aislamiento & purificación , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/aislamiento & purificación , Citotoxicidad Inmunológica/efectos de los fármacos , Citotoxicidad Inmunológica/inmunología , Células Dendríticas/efectos de los fármacos , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunoglobulina G , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Ratones Desnudos , Monocitos/inmunología , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/aislamiento & purificación , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
9.
J Immunol ; 172(5): 2845-52, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14978085

RESUMEN

Targeting recycling endocytic receptors with specific Abs provides a means for introducing a variety of tumor-associated Ags into human dendritic cells (DCs), culminating in their efficient presentation to T cells. We have generated a human mAb (B11) against the mannose receptor that is rapidly internalized by DCs through receptor-mediated endocytosis. By genetically linking the melanoma Ag, pmel17, to Ab B11, we obtained the fully human fusion protein, B11-pmel17. Treatment of DCs with B11-pmel17 resulted in the presentation of pmel17 in the context of HLA class I and class II molecules. Thus, potent pmel17-specific T cells were cytotoxic toward gp100(+) HLA-matched melanoma targets, but not HLA-mismatched melanoma or gp100(-) nonmelanoma tumor lines. Importantly, competitive inhibition of lysis of an otherwise susceptible melanoma cell line by cold targets pulsed with known gp100 CD8 T cell epitopes as well as a dose-dependent proliferative response to Th epitopes demonstrates that DCs can process targeted Ag for activation of cytotoxic as well as helper arms of the immune response. Thus, the specific targeting of soluble exogenous tumor Ag to the DC mannose receptor directly contributes to the generation of multiple HLA-restricted Ag-specific T cell responses.


Asunto(s)
Antígenos de Neoplasias/inmunología , Células Dendríticas/inmunología , Marcación de Gen , Antígenos HLA/fisiología , Lectinas Tipo C/inmunología , Lectinas de Unión a Manosa/inmunología , Melanoma/inmunología , Proteínas/inmunología , Receptores de Superficie Celular/inmunología , Linfocitos T Citotóxicos/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/farmacología , Especificidad de Anticuerpos/genética , Antígenos de Neoplasias/genética , Sitios de Unión de Anticuerpos/genética , Unión Competitiva/genética , Unión Competitiva/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular Transformada , Citotoxicidad Inmunológica/genética , Células Dendríticas/citología , Células Dendríticas/metabolismo , Epítopos de Linfocito T/inmunología , Marcación de Gen/métodos , Antígenos HLA/metabolismo , Antígeno HLA-A2/metabolismo , Antígeno HLA-A2/fisiología , Antígenos HLA-D/fisiología , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/genética , Lectinas de Unión a Manosa/metabolismo , Melanoma/prevención & control , Glicoproteínas de Membrana/síntesis química , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas de Neoplasias/síntesis química , Proteínas de Neoplasias/inmunología , Proteínas/genética , Proteínas/fisiología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Proteínas Recombinantes de Fusión/síntesis química , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Transfección , Antígeno gp100 del Melanoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA