Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Immunity ; 55(1): 82-97.e8, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34847356

RESUMEN

CD8+ T cells responding to chronic infection adapt an altered differentiation program that provides some restraint on pathogen replication yet limits immunopathology. This adaptation is imprinted in stem-like cells and propagated to their progeny. Understanding the molecular control of CD8+ T cell differentiation in chronic infection has important therapeutic implications. Here, we find that the chemokine receptor CXCR3 is highly expressed on viral-specific stem-like CD8+ T cells and that one of its ligands, CXCL10, regulates the persistence and heterogeneity of responding CD8+ T cells in spleens of mice chronically infected with lymphocytic choriomeningitis virus. CXCL10 is produced by inflammatory monocytes and fibroblasts of the splenic red pulp, where it grants stem-like cells access to signals promoting differentiation and limits their exposure to pro-survival niches in the white pulp. Consequently, functional CD8+ T cell responses are greater in Cxcl10-/- mice and are associated with a lower viral set point.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Quimiocina CXCL10/metabolismo , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Monocitos/metabolismo , Receptores CXCR3/metabolismo , Bazo/patología , Animales , Antígeno B7-H1/antagonistas & inhibidores , Diferenciación Celular , Proliferación Celular , Autorrenovación de las Células , Quimiocina CXCL10/genética , Enfermedad Crónica , Selección Clonal Mediada por Antígenos , Femenino , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CXCR3/genética
2.
Immunity ; 54(5): 859-874, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33838745

RESUMEN

Chemokines are chemotactic cytokines that regulate the migration of immune cells. Chemokines function as cues for the coordinated recruitment of immune cells into and out of tissue and also guide the spatial organization and cellular interactions of immune cells within tissues. Chemokines are critical in directing immune cell migration necessary to mount and then deliver an effective anti-tumor immune response; however, chemokines also participate in the generation and recruitment of immune cells that contribute to a pro-tumorigenic microenvironment. Here, we review the role of the chemokine system in anti-tumor and pro-tumor immune responses and discuss how malignant cells and the tumor microenvironment regulate the overall chemokine landscape to shape the type and outcome of immune responses to cancer and cancer treatment.


Asunto(s)
Quimiocinas/inmunología , Inmunidad/inmunología , Neoplasias/inmunología , Animales , Carcinogénesis/inmunología , Movimiento Celular/inmunología , Humanos , Microambiente Tumoral/inmunología
3.
Immunity ; 50(6): 1498-1512.e5, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31097342

RESUMEN

Despite compelling rates of durable clinical responses to programmed cell death-1 (PD-1) blockade, advances are needed to extend these benefits to resistant tumors. We found that tumor-bearing mice deficient in the chemokine receptor CXCR3 responded poorly to anti-PD-1 treatment. CXCR3 and its ligand CXCL9 were critical for a productive CD8+ T cell response in tumor-bearing mice treated with anti-PD-1 but were not required for the infiltration of CD8+ T cells into tumors. The anti-PD-1-induced anti-tumor response was facilitated by CXCL9 production from intratumoral CD103+ dendritic cells, suggesting that CXCR3 facilitates dendritic cell-T cell interactions within the tumor microenvironment. CXCR3 ligands in murine tumors and in plasma of melanoma patients were an indicator of clinical response to anti-PD-1, and their induction in non-responsive murine tumors promoted responsiveness to anti-PD-1. Our data suggest that the CXCR3 chemokine system is a biomarker for sensitivity to PD-1 blockade and that augmenting the intratumoral function of this chemokine system could improve clinical outcomes.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Inmunomodulación/efectos de los fármacos , Neoplasias/inmunología , Neoplasias/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Receptores CXCR3/metabolismo , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Biomarcadores , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Epigénesis Genética , Humanos , Activación de Linfocitos , Ratones , Ratones Noqueados , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Nat Immunol ; 15(5): 431-8, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24658051

RESUMEN

CD96, CD226 (DNAM-1) and TIGIT belong to an emerging family of receptors that interact with nectin and nectin-like proteins. CD226 activates natural killer (NK) cell-mediated cytotoxicity, whereas TIGIT reportedly counterbalances CD226. In contrast, the role of CD96, which shares the ligand CD155 with CD226 and TIGIT, has remained unclear. In this study we found that CD96 competed with CD226 for CD155 binding and limited NK cell function by direct inhibition. As a result, Cd96(-/-) mice displayed hyperinflammatory responses to the bacterial product lipopolysaccharide (LPS) and resistance to carcinogenesis and experimental lung metastases. Our data provide the first description, to our knowledge, of the ability of CD96 to negatively control cytokine responses by NK cells. Blocking CD96 may have applications in pathologies in which NK cells are important.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Células Asesinas Naturales/inmunología , Receptores Inmunológicos/metabolismo , Animales , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos T/genética , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Citotoxicidad Inmunológica/genética , Lipopolisacáridos/inmunología , Neoplasias Pulmonares/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nectinas , Metástasis de la Neoplasia , Neoplasias Experimentales/inmunología , Neumonía/inmunología , Unión Proteica/genética , Receptores Virales/metabolismo
5.
Proc Natl Acad Sci U S A ; 110(36): 14711-6, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23964122

RESUMEN

CD73 inhibits antitumor immunity through the activation of adenosine receptors expressed on multiple immune subsets. CD73 also enhances tumor metastasis, although the nature of the immune subsets and adenosine receptor subtypes involved in this process are largely unknown. In this study, we revealed that A2A/A2B receptor antagonists were effective in reducing the metastasis of tumors expressing CD73 endogenously (4T1.2 breast tumors) and when CD73 was ectopically expressed (B16F10 melanoma). A2A(-/-) mice were strongly protected against tumor metastasis, indicating that host A2A receptors enhanced tumor metastasis. A2A blockade enhanced natural killer (NK) cell maturation and cytotoxic function in vitro, reduced metastasis in a perforin-dependent manner, and enhanced NK cell expression of granzyme B in vivo, strongly suggesting that the antimetastatic effect of A2A blockade was due to enhanced NK cell function. Interestingly, A2B blockade had no effect on NK cell cytotoxicity, indicating that an NK cell-independent mechanism also contributed to the increased metastasis of CD73(+) tumors. Our results thus revealed that CD73 promotes tumor metastasis through multiple mechanisms, including suppression of NK cell function. Furthermore, our data strongly suggest that A2A or A2B antagonists may be useful for the treatment of metastatic disease. Overall, our study has potential therapeutic implications given that A2A/A2B receptor antagonists have already entered clinical trials in other therapeutic settings.


Asunto(s)
5'-Nucleotidasa/inmunología , Células Asesinas Naturales/inmunología , Neoplasias Experimentales/inmunología , Receptor de Adenosina A2A/inmunología , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/metabolismo , Antagonistas del Receptor de Adenosina A2/farmacología , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica/efectos de los fármacos , Citotoxicidad Inmunológica/inmunología , Citometría de Flujo , Granzimas/inmunología , Granzimas/metabolismo , Humanos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Pirimidinas/farmacología , Receptor de Adenosina A2A/genética , Receptor de Adenosina A2A/metabolismo , Receptor de Adenosina A2B/inmunología , Receptor de Adenosina A2B/metabolismo , Triazoles/farmacología , Xantinas/farmacología
6.
Eur J Immunol ; 44(7): 2111-20, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24687687

RESUMEN

The NLRP3 inflammasome plays a crucial role in the innate immune response to pathogens and exogenous or endogenous danger signals. Its activity must be precisely and tightly regulated to generate tailored immune responses. However, the immune cell subsets and cytokines controlling NLRP3 inflammasome activity are still poorly understood. Here, we have shown a link between NKT-cell-mediated TNF-α and NLRP3 inflammasome activity. The NLRP3 inflammasome in APCs was critical to potentiate NKT-cell-mediated immune responses, since C57BL/6 NLRP3 inflammasome-deficient mice exhibited reduced responsiveness to α-galactosylceramide. Importantly, NKT cells were found to act as regulators of NLRP3 inflammasome signaling, as NKT-cell-derived TNF-α was required for optimal IL-1ß and IL-18 production by myeloid cells in response to α-galactosylceramide, by acting on the NLRP3 inflammasome priming step. Thus, NKT cells play a role in the positive regulation of NLRP3 inflammasome priming by mediating the production of TNF-α, thus demonstrating another means by which NKT cells control early inflammation.


Asunto(s)
Proteínas Portadoras/fisiología , Inflamación/etiología , Células T Asesinas Naturales/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Células Presentadoras de Antígenos/fisiología , Citocinas/biosíntesis , Galactosilceramidas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR
7.
J Immunol ; 188(8): 3928-39, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22412194

RESUMEN

Attempts to harness mouse type I NKT cells in different therapeutic settings including cancer, infection, and autoimmunity have proven fruitful using the CD1d-binding glycolipid α-galactosylceramide (α-GalCer). In these different models, the effects of α-GalCer mainly relied on the establishment of a type I NKT cell-dependent immune cascade involving dendritic cell, NK cell, B cell, or conventional CD4(+) and CD8(+) T cell activation/regulation as well as immunomodulatory cytokine production. In this study, we showed that γδ T cells, another population of innate-like T lymphocytes, displayed a phenotype of activated cells (cytokine production and cytotoxic properties) and were required to achieve an optimal α-GalCer-induced immune response. Using gene-targeted mice and recombinant cytokines, a critical need for IL-12 and IL-18 has been shown in the α-GalCer-induced IFN-γ production by γδ T cells. Moreover, this cytokine production occurred downstream of type I NKT cell response, suggesting their bystander effect on γδ T cells. In line with this, γδ T cells failed to directly recognize the CD1d/α-GalCer complex. We also provided evidence that γδ T cells increase their cytotoxic properties after α-GalCer injection, resulting in an increase in killing of tumor cell targets. Moreover, using cancer models, we demonstrated that γδ T cells were required for an optimal α-GalCer-mediated anti-tumor activity. Finally, we reported that immunization of wild-type mice with α-GalCer enhanced the adaptive immune response elicited by OVA, and this effect was strongly mediated by γδ T cells. We conclude that γδ T cells amplify the innate and acquired response to α-GalCer, with possibly important outcomes for the therapeutic effects of this compound.


Asunto(s)
Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Inmunidad Adaptativa , Animales , Antígenos CD1d/inmunología , Antígenos CD1d/metabolismo , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/secundario , Citotoxicidad Inmunológica , Galactosilceramidas/farmacología , Inmunidad Innata , Interleucina-12/biosíntesis , Interleucina-12/inmunología , Interleucina-18/biosíntesis , Interleucina-18/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Hígado/citología , Hígado/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Activación de Linfocitos , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/secundario , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Bazo/citología , Bazo/inmunología , Linfocitos T/metabolismo
8.
Semin Cancer Biol ; 22(1): 23-32, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22210181

RESUMEN

Chronic inflammation is a risk factor for tumor development. However, understanding the effect of the immune system on tumor development has only been significantly advanced over the past two decades. We now appreciate that the immune system, in addition to tumor-suppressive function by eliminating nascent transformed tumor cells, can also exert selection pressure on tumor cells and facilitate tumor growth by providing a favorable tumor microenvironment. Yet, the distinctions between tumor-promoting inflammation and tumor-suppressive immunity are still not clear due to the dual role of some cytokines and other molecules in the immune system. The danger signal hypothesis has shaped our view of the role of immunity in cancer development, but still little is known about the exact role of danger signal receptors in cancer progression. In this review, we introduce the processes of cancer immunoediting and inflammation-induced cancer and discuss what is currently known about the role of danger signal receptors in cancer development and progression.


Asunto(s)
Citocinas/fisiología , Vigilancia Inmunológica/fisiología , Inflamasomas/fisiología , Inflamación/complicaciones , Neoplasias/inmunología , Microambiente Tumoral/inmunología , Progresión de la Enfermedad , Proteína HMGB1 , Humanos , Inmunidad Innata , Factores Inmunológicos , Neoplasias/etiología , Receptores Toll-Like
9.
Immunol Cell Biol ; 90(10): 983-6, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23010873

RESUMEN

Asbestos exposure can result in serious and frequently lethal diseases, including malignant mesothelioma. The host sensor for asbestos-induced inflammation is the NLRP3 inflammasome and it is widely assumed that this complex is essential for asbestos-induced cancers. Here, we report that acute interleukin-1ß production and recruitment of immune cells into peritoneal cavity were significantly decreased in the NLRP3-deficient mice after the administration of asbestos. However, NLRP3-deficient mice displayed a similar incidence of malignant mesothelioma and survival times as wild-type mice. Thus, early inflammatory reactions triggered by asbestos are NLRP3-dependent, but NLRP3 is not critical in the chronic development of asbestos-induced mesothelioma. Notably, in a two-stage carcinogenesis-induced papilloma model, NLRP3-deficient mice showed a resistance phenotype in two different strain backgrounds, suggesting a tumour-promoting role of NLRP3 in certain chemically-induced cancer types.


Asunto(s)
Proteínas Portadoras/metabolismo , Mesotelioma/inmunología , Papiloma/inmunología , Neoplasias Pleurales/inmunología , Neoplasias Cutáneas/inmunología , Animales , Amianto/efectos adversos , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Movimiento Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Modelos Animales de Enfermedad , Exposición a Riesgos Ambientales/efectos adversos , Femenino , Humanos , Inflamación/complicaciones , Interleucina-1beta/metabolismo , Mesotelioma/etiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Papiloma/inducido químicamente , Peritoneo/inmunología , Peritoneo/patología , Neoplasias Pleurales/etiología , Neoplasias Cutáneas/inducido químicamente
10.
Immunol Cell Biol ; 89(6): 739-46, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21263463

RESUMEN

Toll-like receptor-4-lipopolysaccharide (LPS)-mediated inflammation is used to delineate signals involved in cross-talk between antigen-presenting cells (APCs) and lymphocytes such as natural killer (NK) cells. Following APC stimulation and cytokine release, NK cells produce interferon (IFN)-γ. High levels of LPS induce endotoxicosis, a systemic inflammatory disease in which IFN-γ causes significant morbidity and mortality. Several studies have highlighted the role of interleukin (IL)-18, IL-1ß, IL-17A and IFN-γ in the development of endotoxicosis, but whether these cytokines interact with each other is yet to be determined. Our data demonstrate that IL-18 and IL-17A have important roles in NK cell IFN-γ production during endotoxicosis. Importantly, we provide the first evidence that IL-18 also has a role in IL-17A production by T-cell receptor (TCR)-δ cells. Furthermore, we demonstrate that IL-18-deficient mice have a defect in γδ T-cell homeostasis and IL-1ß production, both of which can contribute to the development of disease through induction of IL-17A. These results reveal novel requirements for IL-18 in innate immune cell homeostasis and activation, demonstrating that the role of IL-18 in innate immunity occurs at a level other than activation.


Asunto(s)
Interleucina-18/fisiología , Lipopolisacáridos/toxicidad , Animales , Células Cultivadas , Homeostasis/genética , Homeostasis/inmunología , Inflamación/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-17/inmunología , Interleucina-17/metabolismo , Interleucina-18/genética , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/inmunología
11.
Oncoimmunology ; 4(9): e1027468, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26405596

RESUMEN

The Toll-like receptor 3 (TLR3) agonist poly(I:C) is a promising adjuvant for cancer vaccines due to its induction of potent antitumor responses occurring primarily through the activation of dendritic cells (DCs) and natural killer (NK) cells. However, little is known about the role of TLR3 sensing of endogenous ligands in innate tumor immunosurveillance. Here, we investigated whether TLR3 could modulate immune responses and facilitate tumor control without administration of an agonist. We observed only limited impact of TLR3 deficiency on spontaneous carcinogenesis and primary growth of B16F10, E0771 or MC38 tumors when injected subcutaneously to mice. Nevertheless, TLR3 was observed to limit experimental B16F10 lung metastasis, an immunologic constraint dependent on both IFNγ secretion and NK cells. Interestingly, we observed that NK cells derived from Tlr3 null (Tlr3-/- ) mice were hyporesponsive to cytokine stimulation. Indeed, compared with NK cells with intact TLR3, Tlr3-/- NK cells produced significantly reduced pro-inflammatory cytokines, including IFNγ, when incubated in the presence of different combinations of IL-12, IL-18 and IL-15. Bone-marrow chimera experiments established that competent NK cell responses required TLR3 sensing on radio-sensitive immune cells. Intriguingly, although CD8α DCs robustly express high levels of TLR3, we found that those cells were not necessary for efficient IFNγ production by NK cells. Moreover, the defective NK cell phenotype of Tlr3-/- mice appeared to be independent of the gut microbiota. Altogether, our data demonstrate a pivotal role of endogenous TLR3 stimulation for the acquisition of full NK cell functions and immune protection against experimental metastasis.

12.
Cancer Immunol Res ; 2(12): 1125-31, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25480554

RESUMEN

Chemokines are chemotactic cytokines that control the migration of cells between tissues and the positioning and interactions of cells within tissue. The chemokine superfamily consists of approximately 50 endogenous chemokine ligands and 20 G protein-coupled seven-transmembrane spanning signaling receptors. Chemokines mediate the host response to cancer by directing the trafficking of leukocytes into the tumor microenvironment. This migratory response is complex and consists of diverse leukocyte subsets with both antitumor and protumor activities. Although chemokines were initially appreciated as important mediators of immune cell migration, we now know that they also play important roles in the biology of nonimmune cells important for tumor growth and progression. Chemokines can directly modulate the growth of tumors by inducing the proliferation of cancer cells and preventing their apoptosis. They also direct tumor cell movement required for metastasis. Chemokines can also indirectly modulate tumor growth through their effects on tumor stromal cells and by inducing the release of growth and angiogenic factors from cells in the tumor microenvironment. In this Masters of Immunology primer, we focus on recent advances in understanding the complex nature of the chemokine system in tumor biology with a focus on how the chemokine system could be used to augment cancer immunotherapeutic strategies to elicit a more robust and long-lasting host antitumor immune response.


Asunto(s)
Quimiocinas/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Animales , Progresión de la Enfermedad , Humanos , Inmunoterapia , Metástasis de la Neoplasia , Neoplasias/patología , Neoplasias/terapia , Neovascularización Patológica/inmunología , Neovascularización Patológica/metabolismo , Microambiente Tumoral/inmunología
13.
Nat Med ; 20(11): 1301-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25344738

RESUMEN

Some of the anti-neoplastic effects of anthracyclines in mice originate from the induction of innate and T cell-mediated anticancer immune responses. Here we demonstrate that anthracyclines stimulate the rapid production of type I interferons (IFNs) by malignant cells after activation of the endosomal pattern recognition receptor Toll-like receptor 3 (TLR3). By binding to IFN-α and IFN-ß receptors (IFNARs) on neoplastic cells, type I IFNs trigger autocrine and paracrine circuitries that result in the release of chemokine (C-X-C motif) ligand 10 (CXCL10). Tumors lacking Tlr3 or Ifnar failed to respond to chemotherapy unless type I IFN or Cxcl10, respectively, was artificially supplied. Moreover, a type I IFN-related signature predicted clinical responses to anthracycline-based chemotherapy in several independent cohorts of patients with breast carcinoma characterized by poor prognosis. Our data suggest that anthracycline-mediated immune responses mimic those induced by viral pathogens. We surmise that such 'viral mimicry' constitutes a hallmark of successful chemotherapy.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Doxorrubicina/uso terapéutico , Interferón Tipo I/metabolismo , Transducción de Señal , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Antraciclinas/farmacología , Antraciclinas/uso terapéutico , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Quimiocina CXCL10/metabolismo , Doxorrubicina/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunocompetencia/efectos de los fármacos , Interferón Tipo I/biosíntesis , Ratones Endogámicos C57BL , Proteínas de Resistencia a Mixovirus/metabolismo , Terapia Neoadyuvante , Metástasis de la Neoplasia , ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 3/metabolismo , Resultado del Tratamiento
14.
Clin Cancer Res ; 19(20): 5636-46, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23873688

RESUMEN

PURPOSE: To determine the antitumor efficacy and toxicity of a novel combination approach involving adoptive T-cell immunotherapy using chimeric antigen receptor (CAR) T cells with an immunomodulatory reagent for blocking immunosuppression. EXPERIMENTAL DESIGN: We examined whether administration of a PD-1 blocking antibody could increase the therapeutic activity of CAR T cells against two different Her-2(+) tumors. The use of a self-antigen mouse model enabled investigation into the efficacy, mechanism, and toxicity of this combination approach. RESULTS: In this study, we first showed a significant increase in the level of PD-1 expressed on transduced anti-Her-2 CD8(+) T cells following antigen-specific stimulation with PD-L1(+) tumor cells and that markers of activation and proliferation were increased in anti-Her-2 T cells in the presence of anti-PD-1 antibody. In adoptive transfer studies in Her-2 transgenic recipient mice, we showed a significant improvement in growth inhibition of two different Her-2(+) tumors treated with anti-Her-2 T cells in combination with anti-PD-1 antibody. The therapeutic effects observed correlated with increased function of anti-Her-2 T cells following PD-1 blockade. Strikingly, a significant decrease in the percentage of Gr1(+) CD11b(+) myeloid-derived suppressor cells (MDSC) was observed in the tumor microenvironment of mice treated with the combination therapy. Importantly, increased antitumor effects were not associated with any autoimmune pathology in normal tissue expressing Her-2 antigen. CONCLUSION: This study shows that specifically blocking PD-1 immunosuppression can potently enhance CAR T-cell therapy that has significant implications for potentially improving therapeutic outcomes of this approach in patients with cancer.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Neoplasias/genética , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígenos/inmunología , Antineoplásicos/administración & dosificación , Autoinmunidad , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , Células Mieloides/inmunología , Neoplasias/patología , Neoplasias/terapia , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología
15.
Cancer Res ; 72(22): 5721-32, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22986739

RESUMEN

The NLRP3 inflammasome acts as a danger signal sensor that triggers and coordinates the inflammatory response upon infectious insults or tissue injury and damage. However, the role of the NLRP3 inflammasome in natural killer (NK) cell-mediated control of tumor immunity is poorly understood. Here, we show in a model of chemical-induced carcinogenesis and a series of experimental and spontaneous metastases models that mice lacking NLRP3 display significantly reduced tumor burden than control wild-type (WT) mice. The suppression of spontaneous and experimental tumor metastases and methylcholanthrene (MCA)-induced sarcomas in mice deficient for NLRP3 was NK cell and IFN-γ-dependent. Focusing on the amenable B16F10 experimental lung metastases model, we determined that expression of NLRP3 in bone marrow-derived cells was necessary for optimal tumor metastasis. Tumor-driven expansion of CD11b(+)Gr-1(intermediate) (Gr-1(int)) myeloid cells within the lung tumor microenvironment of NLRP3(-/-) mice was coincident with increased lung infiltrating activated NK cells and an enhanced antimetastatic response. The CD11b(+)Gr-1(int) myeloid cells displayed a unique cell surface phenotype and were characterized by their elevated production of CCL5 and CXCL9 chemokines. Adoptive transfer of this population into WT mice enhanced NK cell numbers in, and suppression of, B16F10 lung metastases. Together, these data suggested that NLRP3 is an important suppressor of NK cell-mediated control of carcinogenesis and metastases and identify CD11b(+)Gr-1(int) myeloid cells that promote NK cell antimetastatic function.


Asunto(s)
Proteínas Portadoras/inmunología , Células Asesinas Naturales/inmunología , Neoplasias Experimentales/inmunología , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Antígeno CD11b/inmunología , Quimiocina CCL5/inmunología , Quimiocina CXCL9/inmunología , Fibrosarcoma/inducido químicamente , Fibrosarcoma/inmunología , Fibrosarcoma/patología , Inflamasomas/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Masculino , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/secundario , Metilcolantreno , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Metástasis de la Neoplasia , Neoplasias Experimentales/patología , Neoplasias Experimentales/secundario , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología
16.
Cancer Res ; 72(16): 3906-11, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22751463

RESUMEN

Hypoxia within a tumor acts as a strong selective pressure that promotes angiogenesis, invasion, and metastatic spread. In this study, we used immune competent bone marrow chimeric mice and syngeneic orthotopic mammary cancer models to show that hypoxia in the primary tumor promotes premetastatic niche formation in secondary organs. Injection of mice with cell-free conditioned medium derived from hypoxic mammary tumor cells resulted in increased bone marrow-derived cell infiltration into the lung in the absence of a primary tumor and led to increased metastatic burden in mammary and melanoma experimental metastasis models. By characterizing the composition of infiltrating bone marrow-derived cells, we identified CD11b+/Ly6Cmed/Ly6G+ myeloid and CD3-/NK1.1+ immune cell lineages as key constituents of the premetastatic niche. Furthermore, the cytotoxicity of natural killer (NK) cells was significantly decreased, resulting in a reduced antitumor response that allowed metastasis formation in secondary organs to a similar extent as ablation of NK cells. In contrast, metastatic burden was decreased when active NK cells were present in premetastatic lungs. Together, our findings suggest that primary tumor hypoxia provides cytokines and growth factors capable of creating a premetastatic niche through recruitment of CD11b+/Ly6Cmed/Ly6G+ myeloid cells and a reduction in the cytotoxic effector functions of NK cell populations.


Asunto(s)
Hipoxia de la Célula/inmunología , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/inmunología , Melanoma Experimental/inmunología , Animales , Antígenos Ly/biosíntesis , Antígenos Ly/inmunología , Antígeno CD11b/biosíntesis , Antígeno CD11b/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Femenino , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Lesiones Precancerosas/inmunología , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA