Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Discov Oncol ; 15(1): 381, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39207605

RESUMEN

Basal Cell Adhesion Molecule (BCAM), a receptor for laminin subunit α5, plays a crucial role in the pathogenesis of various malignancies. Notably, evidence of hypermethylation at multiple immune checkpoints in patients with low BCAM expression suggests these individuals may respond favorably to immunotherapy using ICIs (immune checkpoint inhibitors). This finding lays the foundation for the hypothesis that BCAM may serve as an important biomarker in cancer patients. To investigate this potential, we evaluated BCAM expression patterns in 3114 patients from both discovery and validation cohorts, spanning seven cancer types, using quantitative immunofluorescence (QIF). We also explored the correlation between BCAM and PD-L1 expressions within these cohorts, aiming to establish its potential predictive value for immunotherapy response. Our findings indicate that BCAM was highly expressed in ovarian (79.2%) and lung (78.5%) tumors, with lower yet significant expression in breast (37.7%), head and neck (31.3%), and bladder-urothelial tumors (27.6%). Notably, high BCAM expression was associated with better OS in NSCLC. More importantly, BCAM expression did not correlate with PD-L1 protein expression in any of these tumors, highlighting its independent predictive potential. The widespread expression of BCAM across multiple tumor types, coupled with its lack of correlation with PD-L1 expression, highlights its potential as a predictive novel biomarker across various cancer types.

2.
Sci Immunol ; 9(94): eadh2334, 2024 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-38669316

RESUMEN

T cells are often absent from human cancer tissues during both spontaneously induced immunity and therapeutic immunotherapy, even in the presence of a functional T cell-recruiting chemokine system, suggesting the existence of T cell exclusion mechanisms that impair infiltration. Using a genome-wide in vitro screening platform, we identified a role for phospholipase A2 group 10 (PLA2G10) protein in T cell exclusion. PLA2G10 up-regulation is widespread in human cancers and is associated with poor T cell infiltration in tumor tissues. PLA2G10 overexpression in immunogenic mouse tumors excluded T cells from infiltration, resulting in resistance to anti-PD-1 immunotherapy. PLA2G10 can hydrolyze phospholipids into small lipid metabolites, thus inhibiting chemokine-mediated T cell mobility. Ablation of PLA2G10's enzymatic activity enhanced T cell infiltration and sensitized PLA2G10-overexpressing tumors to immunotherapies. Our study implicates a role for PLA2G10 in T cell exclusion from tumors and suggests a potential target for cancer immunotherapy.


Asunto(s)
Neoplasias , Linfocitos T , Regulación hacia Arriba , Animales , Femenino , Humanos , Ratones , Línea Celular Tumoral , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos C57BL , Neoplasias/inmunología , Fosfolipasas A/inmunología , Fosfolipasas A/genética , Fosfolipasas A2/inmunología , Linfocitos T/inmunología , Regulación hacia Arriba/inmunología
3.
Sci Adv ; 10(9): eadj4698, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38427724

RESUMEN

Cancers exploit coinhibitory receptors on T cells to escape tumor immunity, and targeting such mechanisms has shown remarkable clinical benefit, but in a limited subset of patients. We hypothesized that cancer cells mimic noncanonical mechanisms of early development such as axon guidance pathways to evade T cell immunity. Using gain-of-function genetic screens, we profiled axon guidance proteins on human T cells and their cognate ligands and identified fibronectin leucine-rich transmembrane protein 3 (FLRT3) as a ligand that inhibits T cell activity. We demonstrated that FLRT3 inhibits T cells through UNC5B, an axon guidance receptor that is up-regulated on activated human T cells. FLRT3 expressed in human cancers favored tumor growth and inhibited CAR-T and BiTE + T cell killing and infiltration in humanized cancer models. An FLRT3 monoclonal antibody that blocked FLRT3-UNC5B interactions reversed these effects in an immune-dependent manner. This study supports the concept that axon guidance proteins mimic T cell checkpoints and can be targeted for cancer immunotherapy.


Asunto(s)
Neoplasias , Linfocitos T , Humanos , Neoplasias/genética , Neoplasias/terapia , Inmunoterapia , Glicoproteínas de Membrana , Receptores de Netrina
4.
J Clin Invest ; 133(22)2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37966113

RESUMEN

Effective eradication of leukemic stem cells (LSCs) remains the greatest challenge in treating acute myeloid leukemia (AML). The immune receptor LAIR-1 has been shown to regulate LSC survival; however, the therapeutic potential of this pathway remains unexplored. We developed a therapeutic LAIR-1 agonist antibody, NC525, that induced cell death of LSCs, but not healthy hematopoietic stem cells in vitro, and killed LSCs and AML blasts in both cell- and patient-derived xenograft models. We showed that LAIR-1 agonism drives a unique apoptotic signaling program in leukemic cells that was enhanced in the presence of collagen. NC525 also significantly improved the activity of azacitidine and venetoclax to establish LAIR-1 targeting as a therapeutic strategy for AML that may synergize with standard-of-care therapies.


Asunto(s)
Leucemia Mieloide Aguda , Animales , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Células Madre Hematopoyéticas/metabolismo , Transducción de Señal , Modelos Animales de Enfermedad , Células Madre Neoplásicas/metabolismo
5.
Front Immunol ; 14: 1199513, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37662958

RESUMEN

It has been known for decades that the tumor extracellular matrix (ECM) is dysfunctional leading to loss of tissue architecture and promotion of tumor growth. The altered ECM and tumor fibrogenesis leads to tissue stiffness that act as a physical barrier to immune cell infiltration into the tumor microenvironment (TME). It is becoming increasingly clear that the ECM plays important roles in tumor immune responses. A growing body of data now indicates that ECM components also play a more active role in immune regulation when dysregulated ECM components act as ligands to interact with receptors on immune cells to inhibit immune cell subpopulations in the TME. In addition, immunotherapies such as checkpoint inhibitors that are approved to treat cancer are often hindered by ECM changes. In this review we highlight the ways by which ECM alterations affect and regulate immunity in cancer. More specifically, how collagens and major ECM components, suppress immunity in the complex TME. Finally, we will review how our increased understanding of immune and immunotherapy regulation by the ECM is leading towards novel disruptive strategies to overcome immune suppression.


Asunto(s)
Colágeno , Neoplasias , Humanos , Matriz Extracelular , Inmunoterapia , Terapia de Inmunosupresión , Neoplasias/terapia , Microambiente Tumoral
7.
Cancer Res Commun ; 3(3): 471-482, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36960400

RESUMEN

Targeting the interaction of leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) and its ligands has been shown to reinstate antitumor immunity. In addition, the introduction of the LAIR-1 decoy protein, LAIR-2, sensitizes previously resistant lung tumors to programmed death-1 (PD-1) blockade, indicating the potential of LAIR-1 as an alternative marker for anti-PD-1 resistance in lung cancer. Here, we assessed LAIR-1 as compared with programmed death-ligand 1 (PD-L1) expression in various tumors, with a focus on non-small cell lung cancer (NSCLC) and its histologic subtypes using multiplexed quantitative immunofluorescence (mQIF) in 287 (discovery cohort) and 144 (validation cohort) patients with NSCLC. In addition, using multispectral imaging technology on mQIF images, we evaluated the localization of LAIR-1 on various cell types. We observed that CD14+, CD68+, and CD163+ monocytes and CK+ tumor cells predominantly expressed LAIR-1 more than other cell types. Furthermore, LAIR-1 expression in the tumor compartment was significantly higher in patients with lung adenocarcinoma (LUAD) than those with lung squamous cell carcinoma subtype (**, P = 0.003). Our results indicated that high tumor LAIR-1 expression in patients with LUAD is negatively associated with OS (overall survival, HR = 2.4; *, P = 0.02) highlighting its prognostic value in LUAD but not in other subtypes. The Pearson correlation between LAIR-1 and PD-L1 is 0.31; however, mutual exclusive staining pattern (i.e., several cases were positive for LAIR-1 and negative for PD-L1) was observed. Altogether, our data suggest that the combination therapy of anti-PD-1/PD-L1 with anti-LAIR-1 or the anti-LAIR-1 monotherapy alone may be promising cancer immunotherapeutic strategies. Significance: The spatial, quantitative assessment of LAIR-1 in NSCLC shows positive association of OS with high LAIR-1+/CD68+ cell densities and negative association of OS with high LAIR-1 expression in LUAD tumor subtype.


Asunto(s)
Adenocarcinoma del Pulmón , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Antígeno B7-H1/genética , Leucocitos/metabolismo , Inmunoglobulinas/uso terapéutico
8.
Sci Transl Med ; 11(522)2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31826980

RESUMEN

Systemic lupus erythematosus (SLE) and discoid lupus erythematosus (DLE) of the skin are autoimmune diseases characterized by inappropriate immune responses against self-proteins; the key elements that determine disease pathogenesis and progression are largely unknown. Here, we show that mice lacking immune inhibitory receptor VISTA or programmed death-1 homolog (PD-1H KO) on a BALB/c background spontaneously develop cutaneous and systemic autoimmune diseases resembling human lupus. Cutaneous lupus lesions of PD-1H KO mice have clustering of plasmacytoid dendritic cells (pDCs) similar to human DLE. Using mass cytometry, we identified proinflammatory neutrophils as critical early immune infiltrating cells within cutaneous lupus lesions of PD-1H KO mice. We also found that PD-1H is highly expressed on immune cells in human SLE, DLE lesions, and cutaneous lesions of MRL/lpr mice. A PD-1H agonistic monoclonal antibody in MRL/lpr mice reduces cutaneous disease, autoantibodies, inflammatory cytokines, chemokines, and immune cell expansion. Furthermore, PD-1H on both T cells and myeloid cells including neutrophils and pDCs could transmit inhibitory signals, resulting in reduced activation and function, establishing PD-1H as an inhibitory receptor on T cells and myeloid cells. On the basis of these findings, we propose that PD-1H is a critical element in the pathogenesis and progression of lupus, and PD-1H activation could be effective for treatment of systemic and cutaneous lupus.


Asunto(s)
Autoinmunidad , Lupus Eritematoso Cutáneo/inmunología , Lupus Eritematoso Sistémico/inmunología , Proteínas de la Membrana/metabolismo , Animales , Artritis/patología , Autoanticuerpos/inmunología , Células Dendríticas/inmunología , Humanos , Inflamación/patología , Interferón Tipo I/metabolismo , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/deficiencia , Ratones Endogámicos BALB C , Ratones Endogámicos MRL lpr , Células Mieloides/metabolismo , Neutrófilos/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Terpenos , Regulación hacia Arriba
9.
Nat Med ; 25(4): 656-666, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30833750

RESUMEN

Overexpression of the B7-H1 (PD-L1) molecule in the tumor microenvironment (TME) is a major immune evasion mechanism in some patients with cancer, and antibody blockade of the B7-H1/PD-1 interaction can normalize compromised immunity without excessive side-effects. Using a genome-scale T cell activity array, we identified Siglec-15 as a critical immune suppressor. While only expressed on some myeloid cells normally, Siglec-15 is broadly upregulated on human cancer cells and tumor-infiltrating myeloid cells, and its expression is mutually exclusive to B7-H1, partially due to its induction by macrophage colony-stimulating factor and downregulation by IFN-γ. We demonstrate that Siglec-15 suppresses antigen-specific T cell responses in vitro and in vivo. Genetic ablation or antibody blockade of Siglec-15 amplifies anti-tumor immunity in the TME and inhibits tumor growth in some mouse models. Taken together, our results support Siglec-15 as a potential target for normalization cancer immunotherapy.


Asunto(s)
Inmunoglobulinas/metabolismo , Inmunoterapia , Proteínas de la Membrana/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Animales , Línea Celular Tumoral , Proliferación Celular , Epítopos , Humanos , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Neoplasias/patología , Proteoma/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Linfocitos T/inmunología
10.
Sci Rep ; 7(1): 6086, 2017 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-28729608

RESUMEN

Programmed death one homolog (PD-1H) is an immunoglobulin superfamily molecule and primarily acts as a coinhibitor in the initiation of T cell response to antigens. Here, we report that genetic ablation of PD-1H in mice blocks the differentiation of naive T cells to Foxp3+ inducible Treg cells (iTreg) with a significant decrease of iTreg in lymphoid organs. This effect of PD-1H is highly specific for iTreg because both naturally generated iTreg in gut-related tissues and in vitro induced iTreg by TGF-ß were decreased whereas the genesis of natural Treg (nTreg) remains normal. The suppressive function of both iTreg and nTreg, however, is not affected by the loss of PD-1H. In addition to decreased production, PD-1H deficient iTreg could also rapidly convert to CD4+ T helper 1 or T helper 17 cells in an inflammatory environment. Our results indicate that PD-1H is required for maintenance of iTreg pool size by promoting its differentiation and preventing its conversion to other CD4+ T cell subsets. These findings may have important implications for manipulating Tregs to control inflammation.


Asunto(s)
Diferenciación Celular/genética , Proteínas de la Membrana/genética , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Animales , Apoptosis/genética , Biomarcadores , Citocinas/metabolismo , Elementos de Facilitación Genéticos , Factores de Transcripción Forkhead/genética , Tolerancia Inmunológica , Mediadores de Inflamación/metabolismo , Metilación , Ratones , Ratones Noqueados , Factor de Transcripción STAT3/metabolismo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Células Th17/citología , Células Th17/inmunología , Células Th17/metabolismo
11.
Oncoimmunology ; 5(8): e1185583, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27622059

RESUMEN

Although immune infiltrates in ovarian cancer are associated with improved survival, the ovarian tumor environment has been characterized as immunosuppressive, due in part to functional shifts among dendritic cells with disease progression. We hypothesized that flux in dendritic cell subpopulations with cancer progression were responsible for observed differences in antitumor immune responses in early and late-stage disease. Here we identify three dendritic cell subsets with disparate functions in the ovarian tumor environment. CD11c+CD11b(-)CD103(+) dendritic cells are absent in the peritoneal cavity of healthy mice but comprise up to 40% of dendritic cells in tumor-bearing mice and retain T cell stimulatory capacity in advanced disease. Among CD11c+CD11b+ cells, Lair-1 expression distinguishes stimulatory and immunoregulatory DC subsets, which are also enriched in the tumor environment. Notably, PD-L1 is expressed by Lair-1(hi) immunoregulatory dendritic cells, and may contribute to local tumor antigen-specific T cell dysfunction. Using an adoptive transfer model, we find that PD-1 blockade enables tumor-associated CD103(+) dendritic cells to promote disease clearance. These data demonstrate that antitumor immune capacity is maintained among local dendritic cell subpopulations in the tumor environment with cancer progression. Similar dendritic cell subsets are present in malignant ascites from women with ovarian cancer, supporting the translational relevance of these results.

12.
Cancer Immunol Res ; 3(11): 1257-68, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26138335

RESUMEN

Immune checkpoint blockade has shown significant therapeutic efficacy in melanoma and other solid tumors, but results in ovarian cancer have been limited. With evidence that tumor immunogenicity modulates the response to checkpoint blockade, and data indicating that BRCA-deficient ovarian cancers express higher levels of immune response genes, we hypothesized that BRCA(-) ovarian tumors would be vulnerable to checkpoint blockade. To test this hypothesis, we used an immunocompetent BRCA1-deficient murine ovarian cancer model to compare treatment with CTLA-4 or PD-1/PD-L1 antibodies alone or combined with targeted cytotoxic therapy using a PARP inhibitor. Correlative studies were performed in vitro using human BRCA1(-) cells. We found that CTLA-4 antibody, but not PD-1/PD-L1 blockade, synergized therapeutically with the PARP inhibitor, resulting in immune-mediated tumor clearance and long-term survival in a majority of animals (P < 0.0001). The survival benefit of this combination was T-cell mediated and dependent on increases in local IFNγ production in the peritoneal tumor environment. Evidence of protective immune memory was observed more than 60 days after completion of therapy. Similar increases in the cytotoxic effect of PARP inhibition in the presence of elevated levels of IFNγ in human BRCA1(-) cancer cells support the translational potential of this treatment protocol. These results demonstrate that CTLA-4 blockade combined with PARP inhibition induces protective antitumor immunity and significant survival benefit in the BRCA1(-) tumor model, and support clinical testing of this regimen to improve outcomes for women with hereditary ovarian cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Antígeno CTLA-4/antagonistas & inhibidores , Neoplasias Ováricas/terapia , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Ubiquitina-Proteína Ligasas/deficiencia , Animales , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/administración & dosificación , Terapia Combinada , Citotoxicidad Inmunológica , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Femenino , Humanos , Memoria Inmunológica , Inmunoterapia/métodos , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/administración & dosificación , Subgrupos de Linfocitos T/inmunología , Células Tumorales Cultivadas
13.
J Immunol ; 194(11): 5294-304, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25917101

RESUMEN

PD-1H is a recently identified cell surface coinhibitory molecule of the B7/CD28 immune modulatory gene family. We showed previously that single injection of a PD-1H agonistic mAb protected mice from graft-versus-host disease (GVHD). In this study, we report two distinct mechanisms operate in PD-1H-induced T cell tolerance. First, signaling via PD-1H coinhibitory receptor potently arrests alloreactive donor T cells from activation and expansion in the initiation phase. Second, donor regulatory T cells are subsequently expanded to maintain long-term tolerance and GVHD suppression. Our study reveals the crucial function of PD-1H as a coinhibitory receptor on alloreactive T cells and its function in the regulation of T cell tolerance. Therefore, PD-1H may be a target for the modulation of alloreactive T cells in GVHD and transplantation.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Tolerancia Inmunológica/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/inmunología , Antígeno B7-1/inmunología , Antígenos CD28/inmunología , Activación de Linfocitos/inmunología , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Receptor de Muerte Celular Programada 1/agonistas
14.
J Clin Invest ; 124(5): 1966-75, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24743150

RESUMEN

T cell activation is regulated by the interactions of surface receptors with stimulatory and inhibitory ligands. Programmed death-1 homolog (PD-1H, also called VISTA) is a member of the CD28 family of proteins and has been shown to act as a coinhibitory ligand on APCs that suppress T cell responses. Here, we determined that PD-1H functions as a coinhibitory receptor for CD4⁺ T cells. CD4⁺ T cells in mice lacking PD-1H exhibited a dramatically increased response to antigen stimulation. Furthermore, delivery of a PD-1H-specific agonist mAb directly inhibited CD4⁺ T cell activation both in vitro and in vivo, validating a coinhibitory function of PD-1H. In a murine model of acute hepatitis, administration of a PD-1H agonist mAb suppressed CD4⁺ T cell-mediated acute inflammation. PD-1H-deficient animals were highly resistant to tumor induction in a murine brain glioma model, and depletion of CD4⁺ T cells, but not CD8⁺ T cells, promoted tumor formation. Together, our findings suggest that PD-1H has potential as a target of immune modulation in the treatment of human inflammation and malignancies.


Asunto(s)
Antígenos B7/inmunología , Linfocitos T CD4-Positivos/inmunología , Inmunidad Celular , Activación de Linfocitos , Enfermedad Aguda , Animales , Anticuerpos Monoclonales de Origen Murino/farmacología , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Antígenos B7/antagonistas & inhibidores , Antígenos B7/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Glioma/genética , Glioma/inmunología , Glioma/patología , Hepatitis/genética , Hepatitis/inmunología , Hepatitis/patología , Humanos , Ratones , Ratones Noqueados
15.
Nat Rev Immunol ; 13(4): 227-42, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23470321

RESUMEN

Co-stimulatory and co-inhibitory receptors have a pivotal role in T cell biology, as they determine the functional outcome of T cell receptor (TCR) signalling. The classic definition of T cell co-stimulation continues to evolve through the identification of new co-stimulatory and co-inhibitory receptors, the biochemical characterization of their downstream signalling events and the delineation of their immunological functions. Notably, it has been recently appreciated that co-stimulatory and co-inhibitory receptors display great diversity in expression, structure and function, and that their functions are largely context dependent. Here, we focus on some of these emerging concepts and review the mechanisms through which T cell activation, differentiation and function is controlled by co-stimulatory and co-inhibitory receptors.


Asunto(s)
Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Antígeno B7-1/inmunología , Antígeno B7-1/metabolismo , Antígeno B7-2/inmunología , Antígeno B7-2/metabolismo , Antígenos CD28/inmunología , Antígenos CD28/metabolismo , Humanos , Modelos Inmunológicos , Receptores de Antígenos de Linfocitos T/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Linfocitos T/metabolismo
16.
Prostate ; 73(6): 651-6, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23129407

RESUMEN

BACKGROUND: Prostatitis, a clinical syndrome characterized by pelvic pain and inflammation, is common in adult males. Although several induced and spontaneous murine models of prostatitis have been explored, the role of genetic background on induction has not been well-defined. METHODS: Using a standard methodology for the induction of experimental autoimmune prostatitis (EAP), we investigated both acute and chronic inflammation on several murine genetic backgrounds. RESULTS: In our colony, nonobese diabetic (NOD) mice evinced spontaneous prostatitis that was not augmented by immunization with rat prostate extract (RPE). In contrast, the standard laboratory strain Balb/c developed chronic inflammation in response to RPE immunization. Development of EAP in other strains was variable. CONCLUSIONS: These data suggest that Balb/c mice injected with RPE may provide a useful model for chronic prostatic inflammation.


Asunto(s)
Enfermedades Autoinmunes , Autoinmunidad/genética , Modelos Animales de Enfermedad , Ratones Endogámicos BALB C , Prostatitis , Enfermedad Aguda , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Autoinmunidad/inmunología , Extractos Celulares/inmunología , Extractos Celulares/farmacología , Enfermedad Crónica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Ratones Endogámicos NOD , Ratones Endogámicos NZB , Próstata/inmunología , Próstata/patología , Prostatitis/genética , Prostatitis/inmunología , Prostatitis/patología , Ratas , Ratas Wistar , Especificidad de la Especie
17.
Yale J Biol Med ; 84(4): 409-21, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22180678

RESUMEN

The aim of cancer immunotherapy is to treat malignant disease by inducing or enhancing cancer specific immune responses. With the identification of tumor-associated antigens (TAAs) in the 1990s, cancer immunotherapy research largely focused on inducing immune responses against TAAs but achieved limited success. More recently, the underlying mechanisms and molecular pathways that cancers manipulate to subvert immune-mediated destruction have been identified, including a set of molecules with potent coinhibitory functions. Coinhibitory molecules are expressed on the surface of immune cells, cancer cells, and stromal cells and negatively regulate immune responses to cancer. In particular, one of these ligand-receptor coinhibitory interactions, B7-H1/PD-1, is critical for modulating immune responses to cancer. This knowledge led to the design of revolutionary new immunotherapeutics based on the manipulation of these molecular pathways. Monoclonal antibodies (mAbs) are the primary immunotherapeutic modality used to promote immune function via antagonism or agonism of inhibitory or stimulatory molecular pathways, respectively. Here, we review current knowledge on the function of the B7-H1/PD-1 pathway in mice and humans, its role in the subversion of immune responses in cancer, and clinical evidence that mAb targeting of this pathway results in profound immune anti-cancer effects.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Transducción de Señal , Animales , Antígeno B7-H1/metabolismo , Humanos , Terapia Molecular Dirigida , Receptor de Muerte Celular Programada 1/metabolismo
18.
J Immunol ; 187(4): 1537-41, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21768399

RESUMEN

Upon interaction with B7 homolog 1, programmed death-1 (PD-1) transmits a critical coinhibitory signal to T cells to negatively regulate immune responses. By extensively searching the genomic database with the IgV region of PD-1, we identified a homolog and named it PD-1 homolog (PD-1H). PD-1H is broadly expressed on the cell surface of hematopoietic cells and could be further upregulated on CD4(+) and CD8(+) T cells following activation. We have generated an mAb against PD-1H, which strikingly prevents acute graft-versus-host disease in semi- and fully allogeneic murine models, leading to full chimerism following treatment. Graft-versus-host disease remains a primary hindrance to successful allogeneic hematopoietic cell transplantation therapy for the treatment of hematologic malignancy. Therefore, manipulation of PD-1H function may provide a new modality for controlling T cell responses to allogeneic tissues in transplant medicine.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Antígenos de Superficie/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas , Animales , Antígenos de Superficie/genética , Proteínas Reguladoras de la Apoptosis/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/terapia , Humanos , Ratones , Receptor de Muerte Celular Programada 1 , Trasplante Homólogo
20.
J Immunother ; 30(3): 251-60, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17414316

RESUMEN

B7-H1, B7-DC, B7-H2, B7-H3, and B7-H4, all new additions to the B7 family, here termed "the new B7s," are emerging as important tools in directing immune function; each with unique, yet often overlapping functions. Clearly, each B7 molecule has developed its own indispensable niche in the immune system. The expression of both stimulatory and inhibitory B7 molecules seems to play an essential role in regulating the immune response to transformed cells through a variety of mechanisms. As specific niches of B7 family members continue to be dissected, their diagnostic and therapeutic potential becomes ever more apparent. In this review, we will discuss the role of the new B7s in activation and inhibition of antitumor immune responses, their prospects in diagnostics, and also potential and developing immunotherapy protocols.


Asunto(s)
Antígeno B7-1/fisiología , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Antígeno B7-1/análisis , Antígeno B7-1/metabolismo , Humanos , Inmunidad , Células del Estroma/química , Células del Estroma/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA