Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 21(10): 1205-1218, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32839608

RESUMEN

Immune-modulating therapies have revolutionized the treatment of chronic diseases, particularly cancer. However, their success is restricted and there is a need to identify new therapeutic targets. Here, we show that natural killer cell granule protein 7 (NKG7) is a regulator of lymphocyte granule exocytosis and downstream inflammation in a broad range of diseases. NKG7 expressed by CD4+ and CD8+ T cells played key roles in promoting inflammation during visceral leishmaniasis and malaria-two important parasitic diseases. Additionally, NKG7 expressed by natural killer cells was critical for controlling cancer initiation, growth and metastasis. NKG7 function in natural killer and CD8+ T cells was linked with their ability to regulate the translocation of CD107a to the cell surface and kill cellular targets, while NKG7 also had a major impact on CD4+ T cell activation following infection. Thus, we report a novel therapeutic target expressed on a range of immune cells with functions in different immune responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inflamación/inmunología , Células Asesinas Naturales/inmunología , Leishmania donovani/fisiología , Leishmaniasis Visceral/inmunología , Malaria/inmunología , Proteínas de la Membrana/metabolismo , Plasmodium/fisiología , Animales , Células Cultivadas , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Exocitosis , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Vesículas Secretoras/metabolismo
4.
Nat Immunol ; 18(9): 1004-1015, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28759001

RESUMEN

Avoiding destruction by immune cells is a hallmark of cancer, yet how tumors ultimately evade control by natural killer (NK) cells remains incompletely defined. Using global transcriptomic and flow-cytometry analyses and genetically engineered mouse models, we identified the cytokine-TGF-ß-signaling-dependent conversion of NK cells (CD49a-CD49b+Eomes+) into intermediate type 1 innate lymphoid cell (intILC1) (CD49a+CD49b+Eomes+) populations and ILC1 (CD49a+CD49b-Eomesint) populations in the tumor microenvironment. Strikingly, intILC1s and ILC1s were unable to control local tumor growth and metastasis, whereas NK cells favored tumor immunosurveillance. Experiments with an antibody that neutralizes the cytokine TNF suggested that escape from the innate immune system was partially mediated by TNF-producing ILC1s. Our findings provide new insight into the plasticity of group 1 ILCs in the tumor microenvironment and suggest that the TGF-ß-driven conversion of NK cells into ILC1s is a previously unknown mechanism by which tumors escape surveillance by the innate immune system.


Asunto(s)
Reprogramación Celular/inmunología , Fibrosarcoma/inmunología , Neoplasias Gastrointestinales/inmunología , Tumores del Estroma Gastrointestinal/inmunología , Inmunidad Innata/inmunología , Células Asesinas Naturales/inmunología , Neoplasias Experimentales/inmunología , Escape del Tumor/inmunología , Animales , Estudios de Casos y Controles , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Células Asesinas Naturales/citología , Linfocitos/citología , Linfocitos/inmunología , Ratones , Análisis de Secuencia de ARN , Transducción de Señal , Factor de Crecimiento Transformador beta/inmunología
5.
Immunity ; 47(4): 789-802.e9, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-29045907

RESUMEN

Inhibitors of the receptor tyrosine kinase c-MET are currently used in the clinic to target oncogenic signaling in tumor cells. We found that concomitant c-MET inhibition promoted adoptive T cell transfer and checkpoint immunotherapies in murine cancer models by increasing effector T cell infiltration in tumors. This therapeutic effect was independent of tumor cell-intrinsic c-MET dependence. Mechanistically, c-MET inhibition impaired the reactive mobilization and recruitment of neutrophils into tumors and draining lymph nodes in response to cytotoxic immunotherapies. In the absence of c-MET inhibition, neutrophils recruited to T cell-inflamed microenvironments rapidly acquired immunosuppressive properties, restraining T cell expansion and effector functions. In cancer patients, high serum levels of the c-MET ligand HGF correlated with increasing neutrophil counts and poor responses to checkpoint blockade therapies. Our findings reveal a role for the HGF/c-MET pathway in neutrophil recruitment and function and suggest that c-MET inhibitor co-treatment may improve responses to cancer immunotherapy in settings beyond c-MET-dependent tumors.


Asunto(s)
Inmunoterapia/métodos , Neoplasias Experimentales/terapia , Neutrófilos/inmunología , Proteínas Proto-Oncogénicas c-met/inmunología , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Estimación de Kaplan-Meier , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Neutrófilos/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
6.
J Transl Med ; 19(1): 278, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34193182

RESUMEN

Advances in immune checkpoint therapy and targeted therapy have led to improvement in overall survival for patients with advanced melanoma. Single agent checkpoint PD-1 blockade and combination with BRAF/MEK targeted therapy demonstrated benefit in overall survival (OS). Superior response rates have been demonstrated with combined PD-1/CTLA-4 blockade, with a significant OS benefit compared with single-agent PD-1 blockade. Despite the progress in diagnosis of melanocytic lesions, correct classification of patients, selection of appropriate adjuvant and systemic therapies, and prediction of response to therapy remain real challenges in melanoma. Improved understanding of the tumor microenvironment, tumor immunity and response to therapy has prompted extensive translational and clinical research in melanoma. Development of novel biomarker platforms may help to improve diagnostics and predictive accuracy for selection of patients for specific treatment. There is a growing evidence that genomic and immune features of pre-treatment tumor biopsies may correlate with response in patients with melanoma and other cancers but they have yet to be fully characterized and implemented clinically. Overall, the progress in melanoma therapeutics and translational research will help to optimize treatment regimens to overcome resistance and develop robust biomarkers to guide clinical decision-making. During the Melanoma Bridge meeting (December 3rd-5th, 2020, Italy) we reviewed the currently approved systemic and local therapies for advanced melanoma and discussed novel biomarker strategies and advances in precision medicine.


Asunto(s)
Inmunoterapia , Melanoma , Humanos , Italia , Melanoma/tratamiento farmacológico , Terapia Molecular Dirigida , Microambiente Tumoral
7.
Blood ; 132(16): 1689-1694, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-29986909

RESUMEN

Immune-based therapies hold promise for the treatment of multiple myeloma (MM), but so far, immune checkpoint blockade targeting programmed cell death protein 1 has not proven effective as single agent in this disease. T-cell immunoglobulin and ITIM domains (TIGIT) is another immune checkpoint receptor known to negatively regulate T-cell functions. In this study, we investigated the therapeutic potential of TIGIT blockade to unleash immune responses against MM. We observed that, in both mice and humans, MM progression was associated with high levels of TIGIT expression on CD8+ T cells. TIGIT+ CD8+ T cells from MM patients exhibited a dysfunctional phenotype characterized by decreased proliferation and inability to produce cytokines in response to anti-CD3/CD28/CD2 or myeloma antigen stimulation. Moreover, when challenged with Vk*MYC mouse MM cells, TIGIT-deficient mice showed decreased serum monoclonal immunoglobulin protein levels associated with reduced tumor burden and prolonged survival, indicating that TIGIT limits antimyeloma immune responses. Importantly, blocking TIGIT using monoclonal antibodies increased the effector function of MM patient CD8+ T cells and suppressed MM development. Altogether our data provide evidence for an immune-inhibitory role of TIGIT in MM and support the development of TIGIT-blocking strategies for the treatment of MM patients.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Linfocitos T CD8-positivos/inmunología , Mieloma Múltiple/prevención & control , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptores Inmunológicos/antagonistas & inhibidores , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Células Cultivadas , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mieloma Múltiple/etiología , Mieloma Múltiple/patología , Receptor de Muerte Celular Programada 1/inmunología , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/fisiología
8.
Semin Cancer Biol ; 48: 91-103, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28467889

RESUMEN

Cancer therapies will increasingly be utilized in combination to treat advanced malignancies so as to increase their long-term efficacy in a greater proportion of patients. In particular, much attention has focused on developing targeted therapies that inhibit the PI3K-AKT-mTOR signaling network which is dysregulated in many cancer types. In addition, there is now a growing appreciation that targeting of these pathways can impact not only on cancer cells, but also host immunity. The clinical success of cancer immunotherapies targeting T-cell immune checkpoint receptors PD-1/PD-L1 has demonstrated the importance of immunoevasion as a hallmark of cancer. In this review, we discuss how PI3K-AKT-mTOR inhibitors target cancer cell biology, attenuate immune cell effector function and modulate the tumor microenvironment. We next discuss how the immunomodulatory potential of these inhibitors can be exploited through rational combinations with immunotherapies and targeted therapies.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/terapia , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Humanos , Terapia Molecular Dirigida , Neoplasias/inmunología , Neoplasias/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Microambiente Tumoral/efectos de los fármacos
9.
Lancet Oncol ; 18(12): e731-e741, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29208439

RESUMEN

Use of immune checkpoint inhibitors targeting the programmed cell death protein-1/programmed cell death-ligand 1 and cytotoxic T lymphocyte-associated protein-4 axes has yielded impressive results in some clinical trials. However, only a subset of patients initially respond to these inhibitors, and increasing clinical evidence indicates that a substantial proportion of initial responders ultimately relapse with lethal, drug-resistant disease months or years later. Studies that have used massively parallel sequencing have shed light on the rich functional landscape of mutations that endow tumour cells with the ability to evade T-cell-mediated immunosurveillance. Cancer genomes bear signatures of clonal evolution and selection, particularly implicating acquired defects in interferon receptor signalling and antigen presentation. In this Review, we discuss the biological processes that operate in the formation of so-called immunoresistant niches, and describe the latest progress in the development of combination strategies to reinstate immunosurveillance in immune-refractory tumours.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Puntos de Control del Ciclo Celular/inmunología , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Anticuerpos Monoclonales/uso terapéutico , Antígeno B7-H1/administración & dosificación , Puntos de Control del Ciclo Celular/efectos de los fármacos , Femenino , Humanos , Masculino , Terapia Molecular Dirigida , Neoplasias/genética , Pronóstico , Receptor de Muerte Celular Programada 1/administración & dosificación , Medición de Riesgo , Resultado del Tratamiento
10.
Blood ; 126(13): 1609-20, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26206951

RESUMEN

IL-17-producing cells are important mediators of graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (SCT). Here we demonstrate that a distinct CD8(+) Tc17 population develops rapidly after SCT but fails to maintain lineage fidelity such that they are unrecognizable in the absence of a fate reporter. Tc17 differentiation is dependent on alloantigen presentation by host dendritic cells (DCs) together with IL-6. Tc17 cells express high levels of multiple prototypic lineage-defining transcription factors (eg, RORγt, T-bet) and cytokines (eg, IL-17A, IL-22, interferon-γ, granulocyte macrophage colony-stimulating factor, IL-13). Targeted depletion of Tc17 early after transplant protects from lethal acute GVHD; however, Tc17 cells are noncytolytic and fail to mediate graft-versus-leukemia (GVL) effects. Thus, the Tc17 differentiation program during GVHD culminates in a highly plastic, hyperinflammatory, poorly cytolytic effector population, which we term "inflammatory iTc17" (iTc17). Because iTc17 cells mediate GVHD without contributing to GVL, therapeutic inhibition of iTc17 development in a clinical setting represents an attractive approach for separating GVHD and GVL.


Asunto(s)
Linfocitos T CD8-positivos/patología , Enfermedad Injerto contra Huésped/patología , Efecto Injerto vs Leucemia , Interleucina-17/inmunología , Trasplante de Células Madre/efectos adversos , Células Th17/patología , Animales , Trasplante de Médula Ósea/efectos adversos , Antígenos CD8/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Enfermedad Injerto contra Huésped/inmunología , Humanos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Th17/inmunología
11.
Trends Immunol ; 34(11): 548-55, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23954142

RESUMEN

Interleukin (IL)-12 and IL-23 share the IL-12p40 molecule. IL-12 promotes T helper (Th)1 immunity and IL-23 promotes Th17 immunity, and it has recently become apparent that the balance between IL-12 and IL-23 is important in carcinogenesis. A series of studies demonstrated that, where tumor initiation, growth, and metastasis are concerned, IL-12 may act independently of interferon (IFN)-γ, and IL-23 independently of IL-17A. This review explores the activity of IL-23 in carcinogenesis. In the context of the tumor-inhibitory effects of IL-12, and tumor-promoting effects of IL-23, we discuss the use of anti-IL-12p/23 monoclonal antibodies (mAbs) in autoimmune inflammatory disorders and the alternative specific neutralization of IL-23.


Asunto(s)
Carcinogénesis/inmunología , Interleucina-12/inmunología , Interleucina-23/inmunología , Neoplasias/inmunología , Neoplasias/patología , Animales , Anticuerpos Monoclonales/inmunología , Enfermedades Autoinmunes/inmunología , Humanos
12.
Nat Rev Immunol ; 5(12): 928-40, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16322746

RESUMEN

Immunotherapy is receiving much attention as a means of treating cancer, but complete, durable responses remain rare for most malignancies. The natural immune system seems to have limitations and deficiencies that might affect its ability to control malignant disease. An alternative to relying on endogenous components in the immune repertoire is to generate lymphocytes with abilities that are greater than those of natural T cells, through genetic modification to produce 'supernatural' T cells. This Review describes how such T cells can circumvent many of the barriers that are inherent in the tumour microenvironment while optimizing T-cell specificity, activation, homing and antitumour function.


Asunto(s)
Vacunas contra el Cáncer , Terapia Genética/métodos , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T/inmunología , Animales , Quimera , Ensayos Clínicos como Asunto , Humanos , Inmunoterapia , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología
13.
Mol Ther ; 22(1): 18-27, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24048441

RESUMEN

The tumor microenvironment can promote tumor growth and reduce treatment efficacy. Tumors can occur in many sites in the body, but how surrounding normal tissues at different anatomical sites affect tumor microenvironments and their subsequent response to therapy is not known.We demonstrated that tumors from renal, colon, or prostate cell lines in orthotopic locations responded to immunotherapy consisting of three agonist antibodies, termed Tri-mAb, to a much lesser extent than the same tumor type located subcutaneously. A tissue-specific response to Tri-mAb was confirmed by ex vivo separation of subcutaneous (SC) or orthotopic tumor cells from stromal cells, followed by reinjection of tumor cells into the opposite site. Compared with SC tumors, orthotopic tumors had a microenvironment associated with a type 2 immune response, related to immunosuppression, and an involvement of alternatively activated macrophages in the kidney model. Orthotopic kidney tumors were more highly vascularized than SC tumors. Neutralizing the macrophage- and Th2-associated molecules chemokine (C-C motif) ligand 2 or interleukin-13 led to a significantly improved therapeutic effect. This study highlights the importance of the tissue of implantation in sculpting the tumor microenvironment. These are important fundamental issues in tumor biology and crucial factors to consider in the design of experimental models and treatment strategies.


Asunto(s)
Inmunoterapia , Neoplasias/inmunología , Neoplasias/patología , Microambiente Tumoral/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Antígenos CD40/antagonistas & inhibidores , Antígenos CD40/inmunología , Línea Celular Tumoral , Quimiocina CCL2/inmunología , Neoplasias del Colon/inmunología , Modelos Animales de Enfermedad , Expresión Génica , Interleucina-13/inmunología , Neoplasias Renales/inmunología , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Neoplasias/mortalidad , Neoplasias/terapia , Neovascularización Patológica/inmunología , Especificidad de Órganos/inmunología , Próstata/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/antagonistas & inhibidores , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Resultado del Tratamiento , 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
14.
Proc Natl Acad Sci U S A ; 108(10): 4141-6, 2011 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-21368108

RESUMEN

Histone deacetylase inhibitors (HDACi) have been successfully used as monotherapies for the treatment of hematological malignancies; however, the single agent effects of HDACi against solid tumors are less robust. Using preclinical models of lymphoma, we have recently demonstrated that HDACi induce tumor cell-specific apoptosis and that this is essential for the therapeutic effects of these agents. Herein, we demonstrate that HDACi can be combined with immune-activating antibodies designed to promote the function of antigen-presenting cells (APCs) and enhance proliferation and survival of cytotoxic T cells (CTL) to stimulate a host antitumor immune response resulting in eradication of established solid tumors. This unique combination therapy was dependent on tumor cell apoptosis mediated by HDACi that stimulated the uptake of dead tumor cells by APCs. Tumor eradication was mediated by CD8(+) CTL that used perforin as the key immune effector molecule. This combination therapy was well tolerated and induced long-term immunological antitumor memory capable of mediating spontaneous tumor eradication upon rechallenge. These studies indicate that the ability of HDACi to mediate subtherapeutic levels of tumor cell apoptosis can be exploited by combining with antibodies that augment host antitumor immune responses to mediate robust and prolonged eradication of solid tumors.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Neoplasias Experimentales/terapia , Animales , Células Presentadoras de Antígenos/inmunología , Terapia Combinada , Ratones , Neoplasias Experimentales/tratamiento farmacológico , Linfocitos T Citotóxicos/inmunología , Vorinostat
15.
Proc Natl Acad Sci U S A ; 108(17): 7142-7, 2011 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-21482773

RESUMEN

Trastuzumab, a monoclonal antibody targeting human epidermal growth factor receptor-2 (HER2/ErbB-2), has become the mainstay of treatment for HER2-positive breast cancer. Nevertheless, its exact mechanism of action has not been fully elucidated. Although several studies suggest that Fc receptor-expressing immune cells are involved in trastuzumab therapy, the relative contribution of lymphocyte-mediated cellular cytotoxicity and antitumor cytokines remains unknown. We report here that anti-ErbB-2 mAb therapy is dependent on the release of type I and type II IFNs but is independent of perforin or FasL. Our study thus challenges the notion that classical antibody-dependent, lymphocyte-mediated cellular cytotoxicity is important for trastuzumab. We demonstrate that anti-ErbB-2 mAb therapy of experimental tumors derived from MMTV-ErbB-2 transgenic mice triggers MyD88-dependent signaling and primes IFN-γ-producing CD8+ T cells. Adoptive cell transfer of purified T cell subsets confirmed the essential role of IFN-γ-producing CD8+ T cells. Notably, anti-ErbB-2 mAb therapy was independent of IL-1R or IL-17Ra signaling. Finally, we investigated whether immunostimulatory approaches with antibodies against programmed death-1 (PD-1) or 41BB (CD137) could be used to capitalize on the immune-mediated effects of trastuzumab. We demonstrate that anti-PD-1 or anti-CD137 mAb can significantly improve the therapeutic activity of anti-ErbB-2 mAb in immunocompetent mice.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos de Superficie , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteínas Reguladoras de la Apoptosis , Interferón Tipo I/inmunología , Interferón gamma/inmunología , Neoplasias Mamarias Animales/tratamiento farmacológico , Receptor ErbB-2/inmunología , Receptores Tipo I de Interleucina-1/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Línea Celular Tumoral , Humanos , Interferón Tipo I/genética , Interferón gamma/genética , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Ratones Transgénicos , Receptor de Muerte Celular Programada 1 , Receptor ErbB-2/genética , Receptores Tipo I de Interleucina-1/genética , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/inmunología , Trastuzumab , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética
16.
Eur J Immunol ; 42(10): 2574-83, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22777705

RESUMEN

IL-17, produced by a distinct lineage of CD4(+) helper T (Th) cells termed Th17 cells, induces the production of pro-inflammatory cytokines from resident cells and it has been demonstrated that over-expression of IL-17 plays a crucial role in the onset of several auto-immune diseases. Here we examined the role of IL-17 in the pathogenesis of autoimmune gastritis, a disease that was previously believed to be mediated by IFN-γ. Significantly higher levels of IL-17 and IFN-γ were found in the stomachs and stomach-draining lymph nodes of mice with severe autoimmune gastritis. Unlike IL-17, which was produced solely by CD4(+) T cells in gastritic mice, the majority of IFN-γ-producing cells were CD8(+) T cells. However, CD8(+) T cells alone were not able to induce autoimmune gastritis. T cells that were deficient in IL-17 or IFN-γ production were able to induce autoimmune gastritis but to a much lower extent compared with the disease induced by wild-type T cells. These data demonstrate that production of neither IL-17 nor IFN-γ by effector T cells is essential for the initiation of autoimmune gastritis, but suggest that both are required for the disease to progress to the late pathogenic stage that includes significant tissue disruption.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Gastritis/inmunología , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Células Th17/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/trasplante , Células Cultivadas , Progresión de la Enfermedad , Interferón gamma/genética , Interleucina-17/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células Th17/trasplante
17.
Proc Natl Acad Sci U S A ; 107(18): 8328-33, 2010 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-20404142

RESUMEN

IL-23 is an important molecular driver of Th17 cells and has strong tumor-promoting proinflammatory activity postulated to occur via adaptive immunity. Conversely, more recently it has been reported that IL-17A elicits a protective inflammation that promotes the activation of tumor-specific CD8(+) T cells. Here we show the much broader impact of IL-23 in antagonizing antitumor immune responses primarily mediated by innate immunity. Furthermore, the majority of this impact was independent of IL-17A, which did not appear critical for many host responses to tumor initiation or metastases. IL-23-deficient mice were resistant to experimental tumor metastases in three models where host NK cells controlled disease. Immunotherapy with IL-2 was more effective in mice lacking IL-23, and again the protection afforded was NK cell mediated and independent of IL-17A. Further investigation revealed that loss of IL-23 promoted perforin and IFN-gamma antitumor effector function in both metastasis models examined. IL-23-deficiency also strikingly protected mice from tumor formation in two distinct mouse models of carcinogenesis where the dependence on host IL-12p40 and IL-17A was quite different. Notably, in the 3'-methylcholanthrene (MCA) induction of fibrosarcoma model, this protection was completely lost in the absence of NK cells. Overall, these data indicate the general role that IL-23 plays in suppressing natural or cytokine-induced innate immunity, promoting tumor development and metastases independently of IL-17A.


Asunto(s)
Inmunidad Innata , Interleucina-17/inmunología , Subunidad p19 de la Interleucina-23/inmunología , Miocardio/inmunología , Neoplasias/inmunología , Neoplasias/patología , Animales , Línea Celular Tumoral , Femenino , Inmunoterapia , Interferón gamma/inmunología , Interleucina-2/inmunología , Subunidad p19 de la Interleucina-23/deficiencia , Células Asesinas Naturales/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia , Perforina/inmunología
18.
Curr Top Microbiol Immunol ; 344: 61-95, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-20512555

RESUMEN

This review specifically examines the role of regulatory T cells (Tregs) in cancer in both mice and the clinic. Due to the rapid refinement of the definition of Tregs and their heterogeneity, emphasis is given to research findings over the past three years. For clarity, this review is broadly divided into three short sections that outline the basic biology of Tregs - (1) Treg lineage and development, (2) Treg subsets, and (3) mechanisms of Treg-mediated immune suppression; followed by two more comprehensive sections that cover; (4) clinical observations of Tregs and cancer, and (5) modifications of Treg biology as cancer immunotherapies. The latter two sections discuss the measurement of function and frequency of Treg in model systems and clinical trials and possible ways to interfere with Treg-mediated immune suppression with the focus on recent pre-clinical and clinical findings.


Asunto(s)
Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Animales , Citotoxicidad Inmunológica , Humanos , Tolerancia Inmunológica , Inmunoterapia , Ratones , Neoplasias/terapia
19.
J Immunol ; 184(10): 5493-501, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20400706

RESUMEN

The blockade of immune suppression against antitumor responses is a particularly attractive strategy when combined with agents that promote tumor-specific CTLs. In this study, we have attempted to further improve the CTL induction and potent antitumor efficacy of a combination mAb-based therapy (termed "trimAb therapy") that comprises tumor cell death-inducing anti-death receptor 5 mAb and immune activating anti-CD40 and anti-CD137 mAbs. Among trimAb-treated tumors, the infiltration of CD4(+) Foxp3(+) cells was greater in progressing tumors compared with stable tumors. Blockade of CTLA-4 (CD152)-mediated signals by an antagonistic mAb substantially increased the tumor rejection rate of trimAb therapy, although the immune responses of draining lymph node cells were not augmented. Interestingly, by comparison, additional treatment with agonistic anti-glucocorticoid-induced TNF receptor mAb, antagonistic anti-programmed death-1 (CD279) mAb, or agonistic anti-OX40 (CD134) mAb significantly augmented immune responses of draining lymph node cells, but did not augment the therapeutic effect of trimAb. CD4 T cell depletion reduced the antitumor effect of anti-CTLA-4 mAb treatment alone, but did not reduce the tumor rejection rate of trimAb in conjunction with anti-CTLA-4 mAb. Thus, the blockade of the CTLA-4-mediated inhibitory signal in tumor infiltrating CTL may be the most effective strategy to augment the effect of immune therapies that generate tumor-specific CTL.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/terapia , Animales , Anticuerpos Bloqueadores/fisiología , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/fisiología , Antígenos CD/inmunología , Antígenos CD/metabolismo , Antígenos de Superficie/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Antígeno CTLA-4 , Citotoxicidad Inmunológica , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Activación de Linfocitos/inmunología , Depleción Linfocítica , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Receptor de Muerte Celular Programada 1 , Transporte de Proteínas/inmunología , Receptores de Factor de Crecimiento Nervioso/inmunología , Receptores OX40/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología
20.
Sci Transl Med ; 14(642): eabj9779, 2022 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-35476594

RESUMEN

Neoadjuvant immunotherapy with anti-cytotoxic T lymphocyte-associated protein 4 (CTLA4) + anti-programmed cell death protein 1 (PD1) monoclonal antibodies has demonstrated remarkable pathological responses and relapse-free survival in ~80% of patients with clinically detectable stage III melanoma. However, about 20% of the treated patients do not respond. In pretreatment biopsies of patients with melanoma, we found that resistance to neoadjuvant CTLA4 + PD1 blockade was associated with a low CD4/interleukin-2 (IL-2) gene signature. Ex vivo, addition of IL-2 to CTLA4 + PD1 blockade induced T cell activation and deep immunological responses in anti-CTLA4 + anti-PD1-resistant human tumor specimens. In the 4T1.2 breast cancer mouse model of neoadjuvant immunotherapy, triple combination of anti-CTLA4 + anti-PD1 + IL-2 cured almost twice as many mice as compared with dual checkpoint inhibitor therapy. This improved efficacy was due to the expansion of tumor-specific CD8+ T cells and improved proinflammatory cytokine polyfunctionality of both CD4+ and CD8+ T effector cells and regulatory T cells. Depletion studies suggested that CD4+ T cells were critical for priming of CD8+ T cell immunity against 4T1.2 and helped in the expansion of tumor-specific CD8+ T cells early after neoadjuvant triple immunotherapy. Our results suggest that the addition of IL-2 can overcome resistance to neoadjuvant anti-CTLA4 + anti-PD1, providing the rationale for testing this combination as a neoadjuvant therapy in patients with early-stage cancer.


Asunto(s)
Antígeno CTLA-4 , Interleucina-2 , Melanoma , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Humanos , Interleucina-2/farmacología , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Melanoma/patología , Ratones , Terapia Neoadyuvante , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA