Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Clin Chim Acta ; 487: 341-348, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30359586

RESUMEN

BACKGROUND: The expression of 4-1BB on peripheral regulatory T cells (Tregs) and conventional T cells (Tconvs) in coronary artery disease (CAD) patients is unknown. We aimed to investigate the expression and clinical correlations of 4-1BB on peripheral Tregs and Tconvs in CAD patients. METHODS: Flow cytometry analysis was used to analyze 4-1BB expression on peripheral Tregs and Tconvs. We compared the percentages of 4-1BB on Tregs and Tconvs in the control (ctrl) group, the stable ischemic heart disease (SIHD) group, and the acute coronary syndrome (ACS) group. The correlations of 4-1BB expression on Tregs and Tconvs with the Gensini score and CRP were examined in the ACS group. The value of 4-1BB percentage on Tregs for predicting CAD in this cardiovascular risk population was also analyzed. RESULTS: A total of 71 participants were enrolled in this study. In all the groups, the percentages of 4-1BB on Tregs were significantly higher than on Tconvs (all P < .05). After adjusting for sex, age, SBP, HbA1c and LDL, 4-1BB percentages on Tregs and Tconvs were significantly higher in the SIHD and ACS groups compared with the ctrl group (all P < .05). The ratio of 4-1BB percentage on Tregs to 4-1BB percentage on Tconvs was higher in the ACS group compared with the ctrl group (P = .010). In the ACS group, CRP was negatively correlated with the Tregs percentage (in CD4+ T cells) and the Tregs percentage to Tconvs percentage ratio. The Gensini score was positively correlated with the 4-1BB percentage on Tregs in the ACS group. Linear regression analysis showed 4-1BB percentage on Tregs independently predicted the Gensini score. Binary logistic regression showed CRP, HbA1c and 4-1BB percentage on Tregs independently predicted the development of CAD (SIHD+ACS) in the whole population. CONCLUSION: 4-1BB expression on peripheral Tregs and Tconvs was increased in SIHD and ACS patients. 4-1BB percentage on Tregs positively correlated with the severity of coronary artery stenosis in ACS patients. 4-1BB percentage on Tregs independently predicted the severity of coronary artery stenosis in an ACS population and development of CAD in a cardiovascular risk population.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Enfermedad de la Arteria Coronaria/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Enfermedad de la Arteria Coronaria/patología , Estudios Transversales , Femenino , Citometría de Flujo , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
2.
J Am Heart Assoc ; 6(3)2017 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-28288971

RESUMEN

BACKGROUND: Excessive angiogenesis is a key feature of vulnerable atherosclerotic plaques, and is considered an independent predictor of cardiovascular risk. CD137 signaling has previously been shown to be involved in atherosclerosis. However, the possible role of CD137 signaling in regulating angiogenesis has not been reported. METHODS AND RESULTS: Apolipoprotein E-deficient (ApoE-/-) mice were used as the in vivo model of atherosclerosis. Masson and immunohistochemical analysis of atherosclerotic plaques and Matrigel plug assay were used to evaluate the angiogenesis. Human umbilical vein endothelial cells and mouse brain microvascular endothelial cells were used as in vitro and ex vivo models to study how CD137 signaling affects angiogenesis. Matrigel tube formation assay, mouse aortic ring angiogenesis assay, and migration and proliferation assay were employed to assess angiogenesis. Western blot was used to detect protein expression. We found increased neovessel formation in atherosclerotic plaques of ApoE-/- mice treated with agonist anti-CD137 antibody. Activation of CD137 signaling induced angiogenesis, endothelial proliferation, and endothelial cell migration. CD137 signaling activates the pro-angiogenic Smad1/5 pathway, induces the phosphorylation of Smad1/5 and nuclear translocation of p-Smad1/5, which in turn promotes the expression and translocation of NFATc1. Blocking CD137 signaling with inhibitory anti-CD137 antibody could inhibit this activation and attenuated agonist anti-CD137 antibody-induced angiogenesis. CONCLUSIONS: These findings suggest that CD137 signaling is a new regulator of angiogenesis by modulating the Smad1/5-NFATc1 pathway.


Asunto(s)
Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/patología , Factores de Transcripción NFATC/genética , Placa Aterosclerótica/genética , Proteína Smad1/genética , Proteína Smad5/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Animales , Aorta Torácica/metabolismo , Aorta Torácica/patología , Apolipoproteínas E/deficiencia , Movimiento Celular , Células Cultivadas , Modelos Animales de Enfermedad , Electroforesis en Gel de Poliacrilamida , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFATC/biosíntesis , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Factores de Riesgo , Proteína Smad1/biosíntesis , Proteína Smad5/biosíntesis , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis
3.
Clin Cancer Res ; 23(3): 707-716, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27496866

RESUMEN

PURPOSE: Cetuximab, an EGFR-specific antibody (mAb), modestly improves clinical outcome in patients with head and neck cancer (HNC). Cetuximab mediates natural killer (NK) cell:dendritic cell (DC) cross-talk by cross-linking FcγRIIIa, which is important for inducing antitumor cellular immunity. Cetuximab-activated NK cells upregulate the costimulatory receptor CD137 (4-1BB), which, when triggered by agonistic mAb urelumab, might enhance NK-cell functions, to promote T-cell-based immunity. EXPERIMENTAL DESIGN: CD137 expression on tumor-infiltrating lymphocytes was evaluated in a prospective cetuximab neoadjuvant trial, and CD137 stimulation was evaluated in a phase Ib trial, in combining agonistic urelumab with cetuximab. Flow cytometry and cytokine release assays using NK cells and DC were used in vitro, testing the addition of urelumab to cetuximab-activated NK, DC, and cross presentation to T cells. RESULTS: CD137 agonist mAb urelumab enhanced cetuximab-activated NK-cell survival, DC maturation, and tumor antigen cross-presentation. Urelumab boosted DC maturation markers, CD86 and HLA DR, and antigen-processing machinery (APM) components TAP1/2, leading to increased tumor antigen cross-presentation. In neoadjuvant cetuximab-treated patients with HNC, upregulation of CD137 by intratumoral, cetuximab-activated NK cells correlated with FcγRIIIa V/F polymorphism and predicted clinical response. Moreover, immune biomarker modulation was observed in an open label, phase Ib clinical trial, of patients with HNC treated with cetuximab plus urelumab. CONCLUSIONS: These results suggest a beneficial effect of combination immunotherapy using cetuximab and CD137 agonist in HNC. Clin Cancer Res; 23(3); 707-16. ©2016 AACR.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antineoplásicos Inmunológicos/farmacología , Carcinoma de Células Escamosas/inmunología , Cetuximab/farmacología , Células Dendríticas/efectos de los fármacos , Neoplasias de Cabeza y Cuello/inmunología , Células Asesinas Naturales/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Ligando 4-1BB/inmunología , Anticuerpos Monoclonales/farmacología , Presentación de Antígeno , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Genotipo , Neoplasias de Cabeza y Cuello/patología , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Polimorfismo Genético , Receptor Cross-Talk/efectos de los fármacos , Receptores de IgG/genética , Receptores de IgG/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Regulación hacia Arriba/efectos de los fármacos
4.
Cancer Cell ; 28(4): 415-428, 2015 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-26461090

RESUMEN

T cell engineering is a powerful means to rapidly generate anti-tumor T cells. The costimulatory properties of second-generation chimeric antigen receptors (CARs) determine the overall potency of adoptively transferred T cells. Using an in vivo "stress test" to challenge CD19-targeted T cells, we studied the functionality and persistence imparted by seven different CAR structures providing CD28 and/or 4-1BB costimulation. One configuration, which uses two signaling domains (CD28 and CD3ζ) and the 4-1BB ligand, provided the highest therapeutic efficacy, showing balanced tumoricidal function and increased T cell persistence accompanied by an elevated CD8/CD4 ratio and decreased exhaustion. Remarkably, induction of the IRF7/IFNß pathway was required for optimal anti-tumor activity. Thus, 1928z-41BBL T cells possess strikingly potent intrinsic and immunomodulatory qualities.


Asunto(s)
Antígenos CD28/inmunología , Neoplasias Hematológicas/inmunología , Receptores de Antígenos/inmunología , Linfocitos T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Antígenos CD19/inmunología , Antígenos CD19/metabolismo , Antígenos CD28/biosíntesis , Antígenos CD28/genética , Neoplasias Hematológicas/patología , Neoplasias Hematológicas/terapia , Humanos , Inmunoterapia Adoptiva , Factor 7 Regulador del Interferón/metabolismo , Interferón gamma/metabolismo , Cinética , Activación de Linfocitos/inmunología , Receptores de Antígenos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Transducción de Señal , Linfocitos T/patología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética
5.
Cancer Res ; 75(17): 3466-78, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26113085

RESUMEN

A current pressing need in cancer immunology is the development of preclinical model systems that are immunocompetent for the study of human tumors. Here, we report the development of a humanized murine model that can be used to analyze the pharmacodynamics and antitumor properties of immunostimulatory monoclonal antibodies (mAb) in settings where the receptors targeted by the mAbs are expressed. Human lymphocytes transferred into immunodeficient mice underwent activation and redistribution to murine organs, where they exhibited cell-surface expression of hCD137 and hPD-1. Systemic lymphocyte infiltrations resulted in a lethal CD4(+) T cell-mediated disease (xenograft-versus-host disease), which was aggravated when murine subjects were administered clinical-grade anti-hCD137 (urelumab) and anti-hPD-1 (nivolumab). In mice engrafted with human colorectal HT-29 carcinoma cells and allogeneic human peripheral blood mononuclear cells (PBMC), or with a patient-derived gastric carcinoma and PBMCs from the same patient, we found that coadministration of urelumab and nivolumab was sufficient to significantly slow tumor growth. Correlated with this result were increased numbers of activated human T lymphocytes producing IFNγ and decreased numbers of human regulatory T lymphocytes in the tumor xenografts, possibly explaining the efficacy of the therapeutic regimen. Our results offer a proof of concept for the use of humanized mouse models for surrogate efficacy and histology investigations of immune checkpoint drugs and their combinations.


Asunto(s)
Neoplasias Colorrectales/inmunología , Proteínas de Unión al ADN/genética , Subunidad gamma Común de Receptores de Interleucina/genética , Receptor de Muerte Celular Programada 1/biosíntesis , Linfocitos T/efectos de los fármacos , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Animales , Anticuerpos Monoclonales/administración & dosificación , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Proteínas de Unión al ADN/inmunología , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Células HT29 , Humanos , Subunidad gamma Común de Receptores de Interleucina/inmunología , Leucocitos Mononucleares/inmunología , Activación de Linfocitos/inmunología , Ratones , Nivolumab , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/patología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
6.
Nat Med ; 21(6): 581-90, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25939063

RESUMEN

Chimeric antigen receptors (CARs) targeting CD19 have mediated dramatic antitumor responses in hematologic malignancies, but tumor regression has rarely occurred using CARs targeting other antigens. It remains unknown whether the impressive effects of CD19 CARs relate to greater susceptibility of hematologic malignancies to CAR therapies, or superior functionality of the CD19 CAR itself. We show that tonic CAR CD3-ζ phosphorylation, triggered by antigen-independent clustering of CAR single-chain variable fragments, can induce early exhaustion of CAR T cells that limits antitumor efficacy. Such activation is present to varying degrees in all CARs studied, except the highly effective CD19 CAR. We further determine that CD28 costimulation augments, whereas 4-1BB costimulation reduces, exhaustion induced by persistent CAR signaling. Our results provide biological explanations for the antitumor effects of CD19 CARs and for the observations that CD19 CAR T cells incorporating the 4-1BB costimulatory domain are more persistent than those incorporating CD28 in clinical trials.


Asunto(s)
Neoplasias Hematológicas/inmunología , Receptores de Antígenos/inmunología , Linfocitos T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Antígenos CD19/inmunología , Antígenos CD19/metabolismo , Antígenos CD28/inmunología , Antígenos CD28/metabolismo , Línea Celular Tumoral , Neoplasias Hematológicas/patología , Neoplasias Hematológicas/terapia , Humanos , Inmunoterapia Adoptiva , Interleucina-2/inmunología , Activación de Linfocitos/inmunología , Receptores de Antígenos/metabolismo , Linfocitos T/patología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis
7.
Eur J Immunol ; 45(4): 975-87, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25639669

RESUMEN

It is believed that memory CD8(+) T cells are maintained in secondary lymphoid tissues, peripheral tissues, and BM by homeostatic proliferation. Their survival has been shown to be dependent on IL-7, but it is unclear where they acquire it. Here we show that in murine BM, memory CD8(+) T cells individually colocalize with IL-7(+) reticular stromal cells. The T cells are resting in terms of global transcription and do not express markers of activation, for example, 4-1BB (CD137), IL-2, or IFN-γ, despite the expression of CD69 on about 30% of the cells. Ninety-five percent of the memory CD8(+) T cells in BM are in G0 phase of cell cycle and do not express Ki-67. Less than 1% is in S/M/G2 of cell cycle, according to propidium iodide staining. While previous publications have estimated the extent of proliferation of CD8(+) memory T cells on the basis of BrdU incorporation, we show here that BrdU itself induces proliferation of CD8(+) memory T cells. Taken together, the present results suggest that CD8(+) memory T cells are maintained as resting cells in the BM in dedicated niches with their survival conditional on IL-7 receptor signaling.


Asunto(s)
Células de la Médula Ósea/citología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Fase de Descanso del Ciclo Celular/inmunología , Células del Estroma/inmunología , Animales , Antígenos CD/biosíntesis , Antígenos de Diferenciación de Linfocitos T/biosíntesis , Células de la Médula Ósea/inmunología , Proliferación Celular , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Interleucina-7/inmunología , Antígeno Ki-67/biosíntesis , Lectinas Tipo C/biosíntesis , Ratones , Ratones Endogámicos C57BL , Transcripción Genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis
8.
Neuropathol Appl Neurobiol ; 41(2): e56-67, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24606203

RESUMEN

AIMS: The prognosis of patients with malignant gliomas is still dismal despite maximum treatment. Novel therapeutic alternatives targeting tumorigenic pathways are, therefore, demanded. In murine glioma models, targeting of tumour necrosis factor receptor superfamily (TNFRSF) 9 led to complete tumour eradication. Thus, TNFRSF9 might also constitute a promising target in human diffuse gliomas. As there is a lack of data, we aimed to define the expression pattern and cellular source of TNFRSF9 in human gliomas. METHODS: We investigated TNFRSF9 expression in normal human central nervous system (CNS) tissue and glioma specimens using immunohistochemistry, immunofluorescence and Western blotting techniques. RESULTS: Our results show that TNFRSF9 is considerably up-regulated in human gliomas when compared with normal brain tissue. In addition, our data provides evidence for an immune cell-independent de novo expression pattern of TNFRSF9 in mainly non-neoplastic reactive astrocytes and excludes classic immunological cell types, namely lymphocytes and microglia as the source of TNFRSF9. Moreover, TNFRSF9 is predominantly expressed in a perivascular and peritumoural distribution with significantly higher expression in IDH-1 mutant gliomas. CONCLUSIONS: Our findings provide a novel, TNFRSF9-positive, reactive astrocytic phenotype and challenge the therapeutic suitability of TNFRSF9 as a promising target for human gliomas.


Asunto(s)
Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Western Blotting , Neoplasias Encefálicas/patología , Niño , Preescolar , Femenino , Técnica del Anticuerpo Fluorescente , Glioma/patología , Humanos , Inmunohistoquímica , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/análisis , Regulación hacia Arriba , Adulto Joven
9.
Cancer Immunol Immunother ; 63(9): 947-58, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24927849

RESUMEN

Agonistic monoclonal antibodies (mAbs) directed against the co-signaling molecule CD137 (4-1BB) elicit potent anti-tumor immunity in mice. This anti-tumor immunity has traditionally been thought to result from the ability of the Fab portion of anti-CD137 to function as an analog for CD137L. Although binding of CD137 by anti-CD137 mAbs has the potential to cross-link the Fc fragments, enabling Fc engagement of low to moderate affinity Fc gamma receptors (FcγR), the relative import of such Fc-FcγR interactions in mediating anti-CD137 associated anti-tumor immunity is unknown. We studied the ability of a rat anti-mouse CD137 mAb (2A) to mediate the anti-tumor response against the EL4E7 lymphoma in WT and FcγR(-/-) strains. 2A-treated FcRγ(-/-) mice had improved anti-tumor immunity against EL4E7, which could be completely recapitulated in FcγRIII(-/-) animals. These improved anti-tumor responses were associated with increased splenic CD8ß T cell and dendritic cell (DC) populations. Furthermore, there was an increase in the number of DCs expressing high levels of the CD40, CD80, and CD86 molecules that are associated with more effective antigen presentation. Our results demonstrate an unexpected inhibitory role for FcγRIII in the anti-tumor function of anti-CD137 and underscore the need to consider antibody isotype when engineering therapeutic mAbs.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Linfoma/terapia , Receptores de IgG/deficiencia , Receptores de IgG/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Animales , Anticuerpos Inmovilizados/inmunología , Anticuerpos Inmovilizados/metabolismo , Anticuerpos Monoclonales/metabolismo , Femenino , Células HEK293 , Humanos , Linfoma/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Ratas , Receptores de IgG/metabolismo , Transfección , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética
10.
Transpl Immunol ; 30(4): 128-35, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24582729

RESUMEN

Immunologic disparities between minor histocompatibility antigen (mHAg) genes on Y (H-Y) and X (H-X) chromosomes contribute to graft-versus-host disease (GVHD) and graft-versus-leukemia (GVL) effects observed in male recipients of a female donor (FtoM) hematopoietic stem cell transplant (HCT). Using in silico prediction tools, a panel of HLA-A0201 restricted H-Y peptides was synthesized. Expression of CD137 was monitored on CD8(+) T cells after brief stimulation with the H-Y peptides in FtoM HLA-A0201 HCT recipients (N=29), and control groups (HLA-A0201 MtoM [N=18], non-HLA-A0201 FtoM [N=14], and HLA-A0201 female volunteers [N=25]). Specific H-Y responses were significantly greater in HLA-A0201 FtoM than controls. CD8(+) T-cell responses to novel H-Y epitopes were shared among multiple patients, showing marked CD45RA(+)CD27 cytolytic effector profiles. These data represent a proof of concept for our in silico/ex vivo CD8(+) T-cell based approach of prediction and validation of H-Y mHAgs in HCT recipients, which may facilitate prospective studies to identify targets/biomarkers of GVHD/GVL.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Enfermedad Injerto contra Huésped/inmunología , Antígeno H-Y/inmunología , Antígeno HLA-A2/inmunología , Trasplante de Células Madre Hematopoyéticas , Adolescente , Adulto , Anciano , Niño , Femenino , Efecto Injerto vs Leucemia/inmunología , Humanos , Interferón gamma/biosíntesis , Antígenos Comunes de Leucocito/inmunología , Masculino , Persona de Mediana Edad , Receptores de Trasplantes , Trasplante Homólogo , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Adulto Joven
11.
J Immunol ; 191(8): 4121-9, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24026081

RESUMEN

Adoptive transfer of T cells genetically modified to express cancer-specific receptors can mediate impressive tumor regression in terminally ill patients. However, T cell function and persistence over time could be hampered by the activation of inhibitory costimulatory pathways, such as programmed death 1 (PD1)/programmed death ligand 1, leading to T cell exhaustion and providing tumor cells with an escape mechanism from immunosurveillance. In addition, the lack of positive costimulation at the tumor site can further dampen T cell response. Thus, as T cell genetic engineering has become clinically relevant, we aimed at enhancing T cell antitumor activity by genetically diverting T cell-negative costimulatory signals into positive ones using chimeric costimulatory retargeting molecules and which are composed of the PD1 extracellular domain fused to the signaling domains of positive costimulatory molecules such as CD28 and 4-1BB. After characterizing the optimal PD1 chimera, we designed and optimized a tripartite retroviral vector that enables the simultaneous expression of this chimeric molecule in conjunction with a cancer-specific TCR. Human T cells, transduced to express a PD1/28 chimeric molecule, exhibited enhanced cytokine secretion and upregulation of activation markers upon coculture with tumor cells. These engineered cells also proliferated better compared with control cells. Finally, we tested the function of these cells in two xenograft models of human melanoma tumors and show that PD1/28-engineered human T cells demonstrated superior antitumor function. Overall, we propose that engineering T cells with a costimulatory retargeting molecule can enhance their function, which bears important implications for the improvement of T cell immunotherapy.


Asunto(s)
Antígenos CD28/metabolismo , Melanoma/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Antígenos CD28/genética , Línea Celular Tumoral , Proliferación Celular , Embrión de Pollo , Femenino , Ingeniería Genética , Humanos , Inmunoterapia Adoptiva , Activación de Linfocitos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Receptor de Muerte Celular Programada 1/genética , Receptores de Antígenos de Linfocitos T , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Clin Exp Immunol ; 174(1): 179-91, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23750604

RESUMEN

Detection and isolation of viable alloreactive T cells at the single-cell level requires a cell surface marker induced specifically upon T cell receptor activation. In this study, a member of the tumour necrosis factor receptor (TNFR)-family, CD137 (4-1BB) was investigated for its potential to identify the total pool of circulating alloreactive T cells. Optimal conditions for sensitive and specific detection of allogeneic-induced CD137 expression on circulating T cells were established. Thereafter, CD137(+) alloreactive T cells were phenotypically and functionally characterized by multi-parameter flow cytometry. Alloantigen-induced CD137 expression identified both alloreactive CD8(+) T cells (mean ± standard error of the mean: 0·21 ± 0·07%) and alloreactive CD4(+) T cells (0·21 ± 0·05%). CD137(+) alloreactive T cells were detected in different T cell subsets, including naive T cells, but were found preferentially in CD28(+) T cells and not in the terminally differentiated T cell subset. Upon allogeneic (re-)stimulation, the cytokine-producing as well as proliferative capacity of T cells resided mainly within the CD137-expressing fraction. About 10% of the CD137(+) alloreactive T cells produced any combination of interferon (IFN)-γ, interleukin (IL)-2 and TNF-α. Polyfunctional alloreactive T cells, defined by multiple cytokine expression, were observed infrequently. In conclusion, activation-induced CD137 expression is a fast assay allowing for detection and functional analysis of the total alloreactive T cell compartment at the single-cell level by multi-parameter flow cytometry.


Asunto(s)
Citometría de Flujo/métodos , Isoantígenos/fisiología , Activación de Linfocitos/inmunología , Subgrupos de Linfocitos T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Citometría de Flujo/normas , Humanos , Inmunofenotipificación/métodos , Inmunofenotipificación/normas , Prueba de Cultivo Mixto de Linfocitos/métodos , Prueba de Cultivo Mixto de Linfocitos/normas , Depleción Linfocítica , Sensibilidad y Especificidad , Células Madre/citología , Células Madre/inmunología , Células Madre/metabolismo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética
13.
Clin Exp Immunol ; 173(1): 150-60, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23607447

RESUMEN

Bronchiolitis obliterans syndrome (BOS) is associated with lack of immunosuppression of T cell proinflammatory cytokines and increased T cell granzyme B. Repeated antigen-driven proliferation down-regulates T cell CD28. We hypothesized that down-regulation of CD28 and up-regulation of alternate co-stimulatory molecules (CD134, CD137, CD152 and CD154) on T cells may be associated with BOS. Co-stimulatory molecules, granzyme B, perforin and intracellular cytokines were measured by flow cytometry on T cells from stable lung transplant patients (n = 38), patients with BOS (n = 20) and healthy controls (n = 10). There was a significant increase in the percentage of CD4/28(null) and CD8/28(null) T cells producing granzyme B, interferon (IFN)-γ and tumour necrosis factor (TNF)-α in BOS compared with stable patients. Down-regulation of CD28 was associated with steroid resistance and up-regulation of CD134, CD137, CD152 and CD154 on CD4(+) T cells and CD137 and CD152 on CD8(+) T cells. There was a significant correlation between increased CD28(null) /CD137 T cells producing IFN-γ, TNF-α with BOS grade (r = 0·861, P < 0·001 for CD28(null) /CD137 IFN-γ/CD8) and time post-transplant (r = 0·698, P < 0·001 for CD28(null) /CD137 IFN-γ/CD8). BOS is associated with down-regulation of CD28 and up-regulation of alternate co-stimulatory molecules on steroid-resistant peripheral blood proinflammatory CD4(+) and CD8(+) T cells. Therapeutic targeting of alternate co-stimulatory molecules on peripheral blood CD28(null) T cells and monitoring response using these assays may help in the management of patients with BOS.


Asunto(s)
Bronquiolitis Obliterante/inmunología , Receptores Coestimuladores e Inhibidores de Linfocitos T/biosíntesis , Complicaciones Posoperatorias/inmunología , Subgrupos de Linfocitos T/metabolismo , Adulto , Bronquiolitis Obliterante/etiología , Antígenos CD28/análisis , Ligando de CD40/biosíntesis , Ligando de CD40/genética , Antígeno CTLA-4/biosíntesis , Antígeno CTLA-4/genética , Estudios de Casos y Controles , Receptores Coestimuladores e Inhibidores de Linfocitos T/genética , Ciclosporina/uso terapéutico , Femenino , Granzimas/análisis , Humanos , Inmunosupresores/uso terapéutico , Interferón gamma/biosíntesis , Interferón gamma/genética , Trasplante de Pulmón , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Perforina/análisis , Complicaciones Posoperatorias/etiología , Receptores OX40/biosíntesis , Receptores OX40/genética , Subgrupos de Linfocitos T/inmunología , Tacrolimus/uso terapéutico , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Regulación hacia Arriba
14.
J Virol ; 87(5): 2617-27, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23255792

RESUMEN

Little is known concerning immunodominance within the CD4 T-cell response to viral infections and its persistence into long-term memory. We tested CD4 T-cell reactivity against each viral protein in persons immunized with vaccinia virus (VV), either recently or more than 40 years ago, as a model self-limited viral infection. Similar tests were done with persons with herpes simplex virus 1 (HSV-1) infection as a model chronic infection. We used an indirect method capable of counting the CD4 T cells in blood reactive with each individual viral protein. Each person had a clear CD4 T-cell dominance hierarchy. The top four open reading frames accounted for about 40% of CD4 virus-specific T cells. Early and long-term memory CD4 T-cell responses to vaccinia virus were mathematically indistinguishable for antigen breadth and immunodominance. Despite the chronic intermittent presence of HSV-1 antigen, the CD4 T-cell dominance and diversity patterns for HSV-1 were identical to those observed for vaccinia virus. The immunodominant CD4 T-cell antigens included both long proteins abundantly present in virions and shorter, nonstructural proteins. Limited epitope level and direct ex vivo data were also consistent with pronounced CD4 T-cell immunodominance. We conclude that human memory CD4 T-cell responses show a pattern of pronounced immunodominance for both chronic and self-limited viral infections and that this pattern can persist over several decades in the absence of antigen.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Memoria Inmunológica , Vacuna contra Viruela/inmunología , Virus Vaccinia/inmunología , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Humanos , Inmunidad Celular , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis , Vacunas Atenuadas
15.
Comput Math Methods Med ; 2012: 850754, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22701144

RESUMEN

We present a mathematical model developed to reproduce the immune response entitled with the combined administration of activated OT1 cytotoxic T lymphocytes (CTLs) and Anti-CD137 monoclonal antibodies. The treatment is directed against melanoma in B16 OVA mouse models exposed to a specific immunotherapy strategy. We model two compartments: the injection point compartment where the treatment is administered and the skin compartment where melanoma tumor cells proliferate. To model the migration of OT1 CTLs and antibodies from the injection to the skin compartment, we use delay differential equations (DDEs). The outcomes of the mathematical model are in good agreement with the in vivo results. Moreover, sensitivity analysis of the mathematical model underlines the key role of OT1 CTLs and suggests that a possible reduction of the number of injected antibodies should not affect substantially the treatment efficacy.


Asunto(s)
Sistema Inmunológico/fisiología , Melanoma/inmunología , Melanoma/patología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Animales , Anticuerpos Monoclonales/química , Línea Celular Tumoral , Humanos , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Modelos Estadísticos , Modelos Teóricos , Linfocitos T Citotóxicos/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis
16.
FASEB J ; 26(8): 3380-92, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22593548

RESUMEN

CD137/TNFR9/41BB was originally described as a surface molecule present on activated T and NK cells. However, its expression is broader among leukocytes, and it is also detected on hypoxic endothelial cells and inflamed blood vessels, as well as in atherosclerotic lesions. Here, we demonstrate that lymphatic endothelial cells (LECs) up-regulate CD137 expression from undetectable baseline levels on stimulation with TNF-α, LPS, and IL-1ß. CD137 cross-linking with an agonistic mAb results in NF-κB nuclear translocation, followed by up-regulation of VCAM and a 3-fold increase in the production of the chemokine CCL21. Accordingly, there is a 50% increase in CCR7-dependent migration toward conditioned medium from activated LECs on CD137 cross-linking with the agonistic mAb or the natural ligand (CD137L). Such an enhancement of cell migration is also observed with monocyte-derived dendritic cells transmigrating across CD137-activated LEC monolayers. Using explanted human dermal tissue, we found that inflamed skin contains abundant CD137(+) lymphatic vessels and that ex vivo incubation of explanted human dermis with TNF-α induces CD137 expression in lymphatic capillaries. More interestingly, treatment with CD137 agonistic antibody induces CCL21 expression and DC accumulation close to lymphatic vessels. Collectively, our results demonstrate that the inflammatory function of lymphatic vessels can be regulated by CD137.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Dendríticas/citología , Células Endoteliales/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/fisiología , Anticuerpos Monoclonales/farmacología , Quimiocina CCL21/fisiología , Dermatitis/patología , Dermatitis/fisiopatología , Humanos , Inflamación/inmunología , Vasos Linfáticos/metabolismo , FN-kappa B/farmacología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba , Molécula 1 de Adhesión Celular Vascular/biosíntesis
17.
Cancer Res ; 71(13): 4617-27, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21546571

RESUMEN

Human T cells engineered to express a chimeric antigen receptor (CAR) specific for folate receptor-α (FRα) have shown robust antitumor activity against epithelial cancers in vitro but not in the clinic because of their inability to persist and home to tumor in vivo. In this study, CARs were constructed containing a FRα-specific scFv (MOv19) coupled to the T-cell receptor CD3ζ chain signaling module alone (MOv19-ζ) or in combination with the CD137 (4-1BB) costimulatory motif in tandem (MOv19-BBζ). Primary human T cells transduced to express conventional MOv19-ζ or costimulated MOv19-BBζ CARs secreted various proinflammatory cytokines, and exerted cytotoxic function when cocultured with FRα(+) tumor cells in vitro. However, only transfer of human T cells expressing the costimulated MOv19-BBζ CAR mediated tumor regression in immunodeficient mice bearing large, established FRα(+) human cancer. MOv19-BBζ CAR T-cell infusion mediated tumor regression in models of metastatic intraperitoneal, subcutaneous, and lung-involved human ovarian cancer. Importantly, tumor response was associated with the selective survival and tumor localization of human T cells in vivo and was only observed in mice receiving costimulated MOv19-BBζ CAR T cells. T-cell persistence and antitumor activity were primarily antigen-driven; however, antigen-independent CD137 signaling by CAR improved T-cell persistence but not antitumor activity in vivo. Our results show that anti-FRα CAR outfitted with CD137 costimulatory signaling in tandem overcome issues of T-cell persistence and tumor localization that limit the conventional FRα T-cell targeting strategy to provide potent antitumor activity in vivo.


Asunto(s)
Receptor 1 de Folato/inmunología , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Animales , Femenino , Humanos , Fragmentos de Inmunoglobulinas/genética , Fragmentos de Inmunoglobulinas/inmunología , Ratones , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/terapia , Ingeniería de Proteínas , Receptores de Antígenos de Linfocitos T/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cell Tissue Res ; 344(3): 567-76, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21560035

RESUMEN

Tumor necrosis factor receptor subfamily 9 (TNFRSF9) plays a potentially important general role in immune function. Tnfrsf9 gene expression has previously been characterized in late pregnant mouse uterus and placenta. However, little is known about its expression in the uterus during the implantation phase of early pregnancy. We have assessed the levels and localization of Tnfrsf9 expression in the mouse uterus and conceptus during implantation. Relative Tnfrsf9 mRNA levels were significantly higher in implantation than in non-implantation site tissue on days 6.5-8.5 of pregnancy. This increase did not depend on the presence of the conceptus, as mRNA levels were not significantly different between pregnant implantation sites and artificially induced deciduomas. Localization by in situ hybridization revealed a subpopulation of endothelial and uterine natural killer cells expressing Tnfrsf9 in the endometrium during implantation. In the developing conceptus, primary trophoblast giant and ectoplacental cells expressed Tnfrsf9 on days 6.5-8.5, followed by expression in the trophoblast giant cell layers surrounding the conceptus on day 9.5 of pregnancy. Two main splice forms of Tnfrsf9 mRNA exist and encode proteins with distinct biological functions; both mRNA splice forms were present in uterine and conceptus tissues as determined by reverse transcription with the polymerase chain reaction. Thus, both membrane and soluble forms of Tnfrsf9 are expressed in specific cell types of the uterus and conceptus during the progression of implantation in mice and possibly have an important function in this process.


Asunto(s)
Implantación del Embrión/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Útero/fisiología , Animales , Decidua/metabolismo , Decidua/fisiología , Femenino , Expresión Génica , Masculino , Ratones , Ratones Noqueados , Embarazo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Útero/metabolismo
19.
Brain Res ; 1386: 200-8, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21352817

RESUMEN

Inflammation and immunity play a crucial role in the pathogenesis of cerebral vasospasm after subarachnoid hemorrhage (SAH). CD137 is recognized as an independent costimulatory molecule of T cells and activator of monocytes. A growing body of evidence indicates that CD137 is vital for inflammation and immunity. Therefore, this study aimed to investigate the expression of CD137 in the basilar artery in a rat SAH model and to clarify the potential role of CD137 in cerebral vasospasm. A total of 107 rats were randomly divided into four groups: control group; day 3, day 5, and day 7 groups. Day 3, day 5, and day 7 groups were all SAH groups. The animals in SAH groups were subjected to injection of autologous blood into cisterna magna twice on day 0 and day 2 and were sacrificed on days 3, 5, and 7, respectively. Cross-sectional area of basilar artery was measured and the CD137 expression was assessed by quantitative real-time PCR, Western blot and immunohistochemistry. The cross-sectional area of basilar artery was found to be 57,944±5581µm(2) in control group, 26,100±2639µm(2) in day 3, 19,723±2412µm(2) in day 5, and 28,800±2980µm(2) in day 7 group, respectively. The basilar artery exhibited vasospasm after SAH and became more severe on day 5. The elevated mRNA and protein of CD137 were detected after SAH and peaked on day 5. CD137 is increasingly expressed in a parallel time course to the development of cerebral vasospasm in a rat experimental model of SAH. These findings indicate the possible role of CD137 in the pathogenesis of cerebral vasospasm after SAH.


Asunto(s)
Arterias Cerebrales/metabolismo , Hemorragia Subaracnoidea/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Vasoespasmo Intracraneal/metabolismo , Animales , Arterias Cerebrales/patología , Arterias Cerebrales/fisiopatología , Modelos Animales de Enfermedad , Masculino , Proyectos Piloto , Ratas , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/fisiopatología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Regulación hacia Arriba/genética , Vasoespasmo Intracraneal/etiología , Vasoespasmo Intracraneal/fisiopatología , Insuficiencia Vertebrobasilar/etiología , Insuficiencia Vertebrobasilar/metabolismo , Insuficiencia Vertebrobasilar/fisiopatología
20.
Cancer Res ; 71(3): 801-11, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21266358

RESUMEN

Agonist monoclonal antibodies (mAb) to the immune costimulatory molecule CD137, also known as 4-1BB, are presently in clinical trials for cancer treatment on the basis of their costimulatory effects on primed T cells and perhaps other cells of the immune system. Here we provide evidence that CD137 is selectively expressed on the surface of tumor endothelial cells. Hypoxia upregulated CD137 on murine endothelial cells. Treatment of tumor-bearing immunocompromised Rag(-/-) mice with agonist CD137 mAb did not elicit any measurable antiangiogenic effects. In contrast, agonist mAb stimulated tumor endothelial cells, increasing cell surface expression of the adhesion molecules intercellular adhesion molecule (ICAM)-1, vascular cell adhesion molecule (VCAM)-1, and E-selectin. When adoptively transferred into mice, activated T lymphocytes derived from CD137-deficient animals entered more avidly into tumor tissue after treatment with agonist mAb. This effect could be neutralized with anti-ICAM-1 and anti-VCAM-1 blocking antibodies. Thus, stimulation of CD137 not only enhanced T-cell activation but also augmented their trafficking into malignant tissue, through direct actions on the blood vessels that irrigate the tumor. Our findings identify an additional mechanism of action that can explain the immunotherapeutic effects of agonist CD137 antibodies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Células Endoteliales/efectos de los fármacos , Linfocitos T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/inmunología , Línea Celular Tumoral , Células Endoteliales/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/biosíntesis , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...