Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Acta Neuropathol ; 135(4): 569-579, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29299667

RESUMEN

Paraneoplastic cerebellar degenerations with anti-Yo antibodies (Yo-PCD) are rare syndromes caused by an auto-immune response against neuronal antigens (Ags) expressed by tumor cells. However, the mechanisms responsible for such immune tolerance breakdown are unknown. We characterized 26 ovarian carcinomas associated with Yo-PCD for their tumor immune contexture and genetic status of the 2 onconeural Yo-Ags, CDR2 and CDR2L. Yo-PCD tumors differed from the 116 control tumors by more abundant T and B cells infiltration occasionally organized in tertiary lymphoid structures harboring CDR2L protein deposits. Immune cells are mainly in the vicinity of apoptotic tumor cells, revealing tumor immune attack. Moreover, contrary to un-selected ovarian carcinomas, 65% of our Yo-PCD tumors presented at least one somatic mutation in Yo-Ags, with a predominance of missense mutations. Recurrent gains of the CDR2L gene with tumor protein overexpression were also present in 59% of Yo-PCD patients. Overall, each Yo-PCD ovarian carcinomas carried at least one genetic alteration of Yo-Ags. These data demonstrate an association between massive infiltration of Yo-PCD tumors by activated immune effector cells and recurrent gains and/or mutations in autoantigen-encoding genes, suggesting that genetic alterations in tumor cells trigger immune tolerance breakdown and initiation of the auto-immune disease.


Asunto(s)
Autoantígenos/genética , Proteínas del Tejido Nervioso/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Degeneración Cerebelosa Paraneoplásica/genética , Degeneración Cerebelosa Paraneoplásica/inmunología , Linfocitos B/inmunología , Linfocitos B/patología , Carcinoma/genética , Carcinoma/inmunología , Carcinoma/patología , Estudios de Cohortes , Femenino , Expresión Génica , Humanos , Inmunoglobulina G/metabolismo , Persona de Mediana Edad , Mutación , Clasificación del Tumor , Neoplasias Ováricas/patología , Degeneración Cerebelosa Paraneoplásica/patología , Linfocitos T/inmunología , Linfocitos T/patología
2.
Gastroenterology ; 151(2): 311-23, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27132185

RESUMEN

BACKGROUND & AIMS: The liver receives blood from the gastrointestinal tract through the portal vein, and thereby is exposed continuously to dietary antigens and commensal bacteria. Alcoholic liver disease (ALD) is associated with intestinal dysbiosis, increased intestinal permeability, release of microbes into the portal circulation, and increased serum levels and liver deposits of IgA. We characterized B-cell production of IgA in livers of mice at homeostasis, after oral immunization, in a mouse model of ALD and in human liver samples. METHODS: We performed studies with Balb/c and C57BL/6-Ly5.1 mice, as well as transgenic mice (quasimonoclonal, activation-induced [cytidine] deaminase-Cre-tamoxifen-dependent estrogen receptor 2 [ERT2], Blimp-1-green fluorescent protein [GFP]). C57BL/6-Ly5.1 mice were fed chronic plus binge ethanol to create a model of ALD. Some mice also were given repeated injections of FTY720, which prevents egress of IgA-secreting cells from Peyer's patches. We obtained nontumor liver tissues from patients with colorectal carcinoma undergoing surgery for liver metastases or hepatocellular carcinoma. B cells were isolated from mouse and human liver tissues and analyzed by flow cytometry and enzyme-linked ImmunoSpot (ELISpot). In wild-type and transgenic mice, we traced newly generated IgA-secreting cells at steady state and after oral immunization with 4-hydroxy-3-nitrophenylacetyl (NP)-Ficoll or cholera toxin. IgA responses were also evaluated in our model of ALD. RESULTS: Livers of control mice contained proliferative plasmablasts that originated from Peyer's patches and produced IgAs reactive to commensal bacteria. After oral immunization with cholera toxin or a thymus-independent antigen, a substantial number of antigen-specific IgA-secreting cells was found in the liver. Mice fed ethanol had features of hepatitis and increased numbers of IgA-secreting cells in liver, compared with mice given control diets, as well as higher levels of serum IgA and IgA deposits in liver sinusoids. Injection of FTY720 during ethanol feeding reduced liver and serum levels of IgA and IgA deposits in liver and prevented liver injury. Human liver tissues contained a significant proportion of IgA-producing plasma cells that shared phenotypic and functional attributes with those from mouse liver, including reactivity to commensal bacteria. CONCLUSIONS: Based on studies of mice and human liver tissues, we found the liver to be a site of IgA production by B cells, derived from gut-associated lymphoid tissues. These IgAs react with commensal bacteria and oral antigens. Livers from mice with ethanol-induced injury contain increased numbers of IgA-secreting cells and have IgA deposits in sinusoids. IgAs in the liver could mediate clearance of gut-derived antigens that arrive through portal circulation at homeostasis and protect these organs from pathogens.


Asunto(s)
Antígenos/inmunología , Hepatocitos/metabolismo , Inmunoglobulina A Secretora/biosíntesis , Intestinos/inmunología , Hígado/citología , Ganglios Linfáticos Agregados/inmunología , Animales , Linfocitos B/inmunología , Hepatocitos/inmunología , Humanos , Hígado/inmunología , Hígado/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
3.
Sci Immunol ; 7(72): eabl9330, 2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35687697

RESUMEN

Radiotherapy (RT) of colorectal cancer (CRC) can prime adaptive immunity against tumor-associated antigen (TAA)-expressing CRC cells systemically. However, abscopal tumor remissions are extremely rare, and the postirradiation immune escape mechanisms in CRC remain elusive. Here, we found that irradiated CRC cells used ATR-mediated DNA repair signaling pathway to up-regulate both CD47 and PD-L1, which through engagement of SIRPα and PD-1, respectively, prevented phagocytosis by antigen-presenting cells and thereby limited TAA cross-presentation and innate immune activation. This postirradiation CD47 and PD-L1 up-regulation was observed across various human solid tumor cells. Concordantly, rectal cancer patients with poor responses to neoadjuvant RT exhibited significantly elevated postirradiation CD47 levels. The combination of RT, anti-SIRPα, and anti-PD-1 reversed adaptive immune resistance and drove efficient TAA cross-presentation, resulting in robust TAA-specific CD8 T cell priming, functional activation of T effectors, and increased T cell clonality and clonal diversity. We observed significantly higher complete response rates to RT/anti-SIRPα/anti-PD-1 in both irradiated and abscopal tumors and prolonged survival in three distinct murine CRC models, including a cecal orthotopic model. The efficacy of triple combination therapy was STING dependent as knockout animals lost most benefit of adding anti-SIRPα and anti-PD-1 to RT. Despite activation across the myeloid stroma, the enhanced dendritic cell function accounts for most improvements in CD8 T cell priming. These data suggest ATR-mediated CD47 and PD-L1 up-regulation as a key mechanism restraining radiation-induced immune priming. RT combined with SIRPα and PD-1 blockade promotes robust antitumor immune priming, leading to systemic tumor regressions.


Asunto(s)
Antígeno CD47 , Neoplasias Colorrectales , Animales , Antígenos de Neoplasias , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Antígeno B7-H1 , Antígeno CD47/metabolismo , Neoplasias Colorrectales/radioterapia , Humanos , Ratones , Receptor de Muerte Celular Programada 1 , Regulación hacia Arriba
4.
Clin Cancer Res ; 27(11): 3050-3060, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33771853

RESUMEN

PURPOSE: As hypoxia can mediate resistance to immunotherapy, we investigated the safety, tolerability, and efficacy of combining evofosfamide, a prodrug that alleviates hypoxia, with ipilimumab, an immune checkpoint inhibitor, in immunologically "cold" cancers, which are intrinsically insensitive to immunotherapy, as well as in "hot/warm" metastatic cancers that are, atypical of such cancers, resistant to immunotherapy. PATIENTS AND METHODS: In a phase I, 3+3 dose-escalation trial (NCT03098160), evofosfamide (400-640 mg/m2) and ipilimumab (3 mg/kg) were administered in four 3-week cycles. The former was administered on days 1 and 8 of cycles 1-2, while the latter was administered on day 8 of cycles 1-4. Response was assessed using immune-related RECIST and retreatment was allowed, if deemed beneficial, after completion of cycle 4 or at progression. RESULTS: Twenty-two patients were enrolled, of whom 21 were evaluable, encompassing castration-resistant prostate cancer (n = 11), pancreatic cancer (n = 7), immunotherapy-resistant melanoma (n = 2), and human papillomavirus-negative head and neck cancer (n = 1). Drug-related hematologic toxicities, rash, fever, nausea, vomiting, and elevation of liver enzymes were observed in > 10% of patients. The most common drug-related grade 3 adverse event was alanine aminotransferase elevation (33.3%). Two patients discontinued ipilimumab and 4 required evofosfamide deescalation due to toxicity. Of 18 patients with measurable disease at baseline, 3 (16.7%) achieved partial response and 12 (66.7%) achieved stable disease. The best responses were observed at 560 mg/m2 evofosfamide. Preexisting immune gene signatures predicted response to therapy, while hypermetabolic tumors predicted progression. Responders also showed improved peripheral T-cell proliferation and increased intratumoral T-cell infiltration into hypoxia. CONCLUSIONS: No new or unexpected safety signals were observed from combining evofosfamide and ipilimumab, and evidence of therapeutic activity was noted.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Ipilimumab/administración & dosificación , Melanoma/tratamiento farmacológico , Nitroimidazoles/administración & dosificación , Neoplasias Pancreáticas/tratamiento farmacológico , Mostazas de Fosforamida/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Anciano , Femenino , Humanos , Ipilimumab/efectos adversos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Nitroimidazoles/efectos adversos , Mostazas de Fosforamida/efectos adversos , Seguridad , Resultado del Tratamiento
5.
Sci Immunol ; 5(46)2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32303573

RESUMEN

Dendritic cells play a key role in the orchestration of antitumor immune responses. The cDC1 (conventional dendritic cell 1) subset has been shown to be essential for antitumor responses and response to immunotherapy, but its precise role in humans is largely unexplored. Using a multidisciplinary approach, we demonstrate that human cDC1 play an important role in the antitumor immune response through their capacity to produce type III interferon (IFN-λ). By analyzing a large cohort of breast primary tumors and public transcriptomic datasets, we observed specific production of IFN-λ1 by cDC1. In addition, both IFN-λ1 and its receptor were associated with favorable patient outcomes. We show that IFN-III promotes a TH1 microenvironment through increased production of IL-12p70, IFN-γ, and cytotoxic lymphocyte-recruiting chemokines. Last, we showed that engagement of TLR3 is a therapeutic strategy to induce IFN-III production by tumor-associated cDC1. These data provide insight into potential IFN- or cDC1-targeting antitumor therapies.


Asunto(s)
Neoplasias de la Mama/inmunología , Células Dendríticas/inmunología , Interferones/biosíntesis , Neoplasias de la Mama/diagnóstico , Femenino , Humanos , Inmunidad Innata/inmunología , Interferones/inmunología , Interferón lambda
6.
Acta Neuropathol Commun ; 7(1): 38, 2019 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-30857565

RESUMEN

Encephalitis with anti-NMDAR antibodies (NMDAR-E) is a severe autoimmune neurological disorder, defined by a clinical presentation of encephalitis and the presence of IgG targeting the GluN1 subunit of NMDA receptors in the CSF. An underlying ovarian teratoma is commonly associated with this autoimmune disease suggesting a role of the tumor in immunopathogenesis. In this study, we characterized the salient histopathological features of 27 ovarian teratomas associated with NMDAR-E (3 immature and 24 mature teratomas) and 40 controls without associated encephalitis. All but one NMDAR-E-associated teratomas contained a nervous tissue component, while less than 40% of control teratomas did (p < 0.001). GluN1 expression by teratomatous nervous tissue seemed to be more often glial in NMDAR-E teratomas than in control teratomas (73% vs. 29%, p < 0.05). Strikingly, 3 out of 24 NMDAR-E-associated mature teratomas contained neuroglial tissue exhibiting histopathological features of central nervous system neuroglial tumor, while such glioma-like features are exceptionally described in the literature on ovarian teratomas. Moreover, NMDAR-E associated teratomas differed from sporadic ovarian teratomas by consistent and prominent infiltration of the nervous tissue component by immune cells, comprised of T- and B-cells and mature dendritic cells organized in tertiary lymphoid structures, with IgG and IgA deposits and plasma cells in close contact to the neuroglial tissue.These data demonstrate an association between massive infiltration of NMDAR-E-associated teratomas by immune cells and particular glial features of its neuroglial component, suggesting that this glial tissue might be involved in triggering or sustaining the anti-tumor response associated with the auto-immune neurological disease.


Asunto(s)
Encefalitis Antirreceptor N-Metil-D-Aspartato/inmunología , Encefalitis Antirreceptor N-Metil-D-Aspartato/patología , Encéfalo/inmunología , Encéfalo/patología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Teratoma/inmunología , Teratoma/patología , Linfocitos B/inmunología , Linfocitos B/patología , Femenino , Humanos , Clasificación del Tumor/métodos , Linfocitos T/inmunología , Linfocitos T/patología
7.
Methods Mol Biol ; 1845: 205-222, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30141015

RESUMEN

Follicular helper T (Tfh) cells are major components of the humoral immune response due to their pivotal role in germinal center formation and antibody affinity maturation following B-cell isotype switching. This CD4+ T-cell subtype is mainly found in the B-cell zone of secondary lymphoid organs as well as in tertiary lymphoid structures (TLS), which are highly organized structures composed of T and B cells, occasionally found at the invasive margin in the tumor microenvironment.We describe here how to perform immunofluorescence staining of tumor tissue sections and multicolor flow cytometry on tumor cell suspensions to identify and visualize these TLS-associated Tfh cells within the tumor microenvironment of various human cancers. These assays take advantage of combinations of markers and molecules involved in Tfh differentiation and function.


Asunto(s)
Neoplasias/inmunología , Neoplasias/patología , Linfocitos T Colaboradores-Inductores/inmunología , Estructuras Linfoides Terciarias/inmunología , Estructuras Linfoides Terciarias/patología , Microambiente Tumoral , Biomarcadores , Análisis de Datos , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunofenotipificación/métodos , Neoplasias/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo , Estructuras Linfoides Terciarias/metabolismo
8.
J Exp Med ; 211(10): 1969-76, 2014 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-25180061

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by the production of antibodies to self-nucleic acids, immune complex deposition, and tissue inflammation such as glomerulonephritis. Innate recognition of self-DNA and -RNA and the ensuing production of cytokines such as type I interferons (IFNs) contribute to SLE development. Plasmacytoid dendritic cells (pDCs) have been proposed as a source of pathogenic IFN in SLE; however, their net contribution to the disease remains unclear. We addressed this question by reducing gene dosage of the pDC-specific transcription factor E2-2 (Tcf4), which causes a specific impairment of pDC function in otherwise normal animals. We report that global or DC-specific Tcf4 haplodeficiency ameliorated SLE-like disease caused by the overexpression of the endosomal RNA sensor Tlr7. Furthermore, Tcf4 haplodeficiency in the B6.Sle1.Sle3 multigenic model of SLE nearly abolished key disease manifestations including anti-DNA antibody production and glomerulonephritis. Tcf4-haplodeficient SLE-prone animals showed a reduction of the spontaneous germinal center reaction and its associated gene expression signature. These results provide genetic evidence that pDCs are critically involved in SLE pathogenesis and autoantibody production, confirming their potential utility as therapeutic targets in the disease.


Asunto(s)
Células Dendríticas/inmunología , Interferón Tipo I/inmunología , Lupus Eritematoso Sistémico/genética , Animales , Anticuerpos Antinucleares/inmunología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Cruzamientos Genéticos , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Dosificación de Gen/genética , Lupus Eritematoso Sistémico/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Estadísticas no Paramétricas , Receptor Toll-Like 7/metabolismo , Factor de Transcripción 4
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA