Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Immunity ; 53(3): 564-580.e9, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32750334

RESUMEN

Tumor immune escape limits durable responses to T cell therapy. Here, we examined how regulation and function of gene products that provide the target epitopes for CD8+ T cell anti-tumor immunity influence therapeutic efficacy and resistance. We used a CRISPR-Cas9-based method (CRISPitope) in syngeneic melanoma models to fuse the same model CD8+ T cell epitope to the C-termini of different endogenous gene products. Targeting melanosomal proteins or oncogenic CDK4R24C (Cyclin-dependent kinase 4) by adoptive cell transfer (ACT) of the same epitope-specific CD8+ T cells revealed diverse genetic and non-genetic immune escape mechanisms. ACT directed against melanosomal proteins, but not CDK4R24C, promoted melanoma dedifferentiation, and increased myeloid cell infiltration. CDK4R24C antigen persistence was associated with an interferon-high and T-cell-rich tumor microenvironment, allowing for immune checkpoint inhibition as salvage therapy. Thus, the choice of target antigen determines the phenotype and immune contexture of recurrent melanomas, with implications to the design of cancer immunotherapies.


Asunto(s)
Traslado Adoptivo/métodos , Linfocitos T CD8-positivos/trasplante , Epítopos de Linfocito T/inmunología , Melanoma/inmunología , Melanoma/terapia , Escape del Tumor/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Epítopos de Linfocito T/genética , Técnicas de Inactivación de Genes , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones , Ratones Endogámicos C57BL , Células Mieloides/citología , Células Mieloides/inmunología , Microambiente Tumoral/inmunología
2.
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
3.
Oncoimmunology ; 6(6): e1320626, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28680756

RESUMEN

Immune checkpoint inhibitors have significantly improved the treatment of several cancers. T-cell infiltration and the number of neoantigens caused by tumor-specific mutations are correlated to favorable responses in cancers with a high mutation load. Accordingly, checkpoint immunotherapy is thought to be less effective in tumors with low mutation frequencies such as neuroblastoma, a neuroendocrine tumor of early childhood with poor outcome of the high-risk disease group. However, spontaneous regressions and paraneoplastic syndromes seen in neuroblastoma patients suggest substantial immunogenicity. Using an integrative transcriptomic approach, we investigated the molecular characteristics of T-cell infiltration in primary neuroblastomas as an indicator of pre-existing immune responses and potential responsiveness to checkpoint inhibition. Here, we report that a T-cell-poor microenvironment in primary metastatic neuroblastomas is associated with genomic amplification of the MYCN (N-Myc) proto-oncogene. These tumors exhibited lower interferon pathway activity and chemokine expression in line with reduced immune cell infiltration. Importantly, we identified a global role for N-Myc in the suppression of interferon and pro-inflammatory pathways in human and murine neuroblastoma cell lines. N-Myc depletion potently enhanced targeted interferon pathway activation by a small molecule agonist of the cGAS-STING innate immune pathway. This promoted chemokine expression including Cxcl10 and T-cell recruitment in microfluidics migration assays. Hence, our data suggest N-Myc inhibition plus targeted IFN activation as adjuvant strategy to enforce cytotoxic T-cell recruitment in MYCN-amplified neuroblastomas.

4.
Nature ; 507(7490): 109-13, 2014 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-24572365

RESUMEN

Intermittent intense ultraviolet (UV) exposure represents an important aetiological factor in the development of malignant melanoma. The ability of UV radiation to cause tumour-initiating DNA mutations in melanocytes is now firmly established, but how the microenvironmental effects of UV radiation influence melanoma pathogenesis is not fully understood. Here we report that repetitive UV exposure of primary cutaneous melanomas in a genetically engineered mouse model promotes metastatic progression, independent of its tumour-initiating effects. UV irradiation enhanced the expansion of tumour cells along abluminal blood vessel surfaces and increased the number of lung metastases. This effect depended on the recruitment and activation of neutrophils, initiated by the release of high mobility group box 1 (HMGB1) from UV-damaged epidermal keratinocytes and driven by Toll-like receptor 4 (TLR4). The UV-induced neutrophilic inflammatory response stimulated angiogenesis and promoted the ability of melanoma cells to migrate towards endothelial cells and use selective motility cues on their surfaces. Our results not only reveal how UV irradiation of epidermal keratinocytes is sensed by the innate immune system, but also show that the resulting inflammatory response catalyses reciprocal melanoma-endothelial cell interactions leading to perivascular invasion, a phenomenon originally described as angiotropism in human melanomas by histopathologists. Angiotropism represents a hitherto underappreciated mechanism of metastasis that also increases the likelihood of intravasation and haematogenous dissemination. Consistent with our findings, ulcerated primary human melanomas with abundant neutrophils and reactive angiogenesis frequently show angiotropism and a high risk for metastases. Our work indicates that targeting the inflammation-induced phenotypic plasticity of melanoma cells and their association with endothelial cells represent rational strategies to specifically interfere with metastatic progression.


Asunto(s)
Inflamación/etiología , Neoplasias Pulmonares/secundario , Melanoma/irrigación sanguínea , Melanoma/patología , Neoplasias Cutáneas/patología , Quemadura Solar/etiología , Rayos Ultravioleta , Animales , Movimiento Celular/efectos de la radiación , Transformación Celular Neoplásica/efectos de la radiación , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Proteína HMGB1/metabolismo , Inmunidad Innata/efectos de la radiación , Queratinocitos/metabolismo , Queratinocitos/patología , Queratinocitos/efectos de la radiación , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/etiología , Masculino , Melanocitos/patología , Melanocitos/efectos de la radiación , Melanoma/etiología , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/etiología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/etiología , Quemadura Solar/complicaciones , Receptor Toll-Like 4/metabolismo
5.
Clin Cancer Res ; 17(17): 5736-47, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21750202

RESUMEN

PURPOSE: To predict the potential antitumor effect of antigen-specific T cells in melanoma patients, we investigated T-cell effector function in relation to tumor-escape mechanisms. EXPERIMENTAL DESIGN: CD8(+) T cells isolated from tumor, adjacent normal skin, and peripheral blood of 17 HLA-A2(+) patients with advanced-stage melanoma were analyzed for their antigen specificity and effector function against melanocyte differentiation antigens MART-1, gp100, and tyrosinase by using HLA-A2/peptide tetramers and functional assays. In addition, the presence of tumor-escape mechanisms PD-L1/PD-1 pathway, FoxP3 and loss of HLA or melanocyte differentiation antigens, both required for tumor cell recognition and killing, were studied. RESULTS: Higher percentages of melanocyte antigen-specific CD8(+) T cells were found in the melanoma tissues as compared with adjacent normal skin and peripheral blood. Functional analysis revealed 2 important findings: (i) in 5 of 17 patients, we found cytokine production after specific peptide stimulation by tumor-infiltrating lymphocytes (TIL), not by autologous peripheral blood lymphocytes (PBL); (ii) CD8(+) T cells from 7 of 17 patients did not produce cytokines after specific stimulation, which corresponded with significant loss of tumor HLA-A2 expression. The presence of other tumor-escape mechanisms did not correlate to T-cell function. CONCLUSIONS: Our data show that functional T-cell responses could be missed when only PBL and not TIL are evaluated, emphasizing the importance of TIL analysis for immunomonitoring. Furthermore, loss of tumor HLA-A2 may explain the lack of T-cell functionality. These findings have important implications for selecting melanoma patients who may benefit from immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunoterapia , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/inmunología , Piel/inmunología , Anciano , Anciano de 80 o más Años , Antígeno B7-H1/biosíntesis , Linfocitos T CD4-Positivos , Células Cultivadas , Citocinas/biosíntesis , Citotoxicidad Inmunológica , Femenino , Factores de Transcripción Forkhead/biosíntesis , Antígeno HLA-A2/biosíntesis , Antígeno HLA-A2/inmunología , Humanos , Activación de Linfocitos , Antígeno MART-1/inmunología , Masculino , Melanoma/sangre , Melanoma/patología , Melanoma/terapia , Persona de Mediana Edad , Monofenol Monooxigenasa/inmunología , Escape del Tumor , Antígeno gp100 del Melanoma/inmunología
6.
J Invest Dermatol ; 131(6): 1240-51, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21326294

RESUMEN

In this study, we report the previously unknown mechanism of inducing robust anti-melanoma immunity by the vitiligo-inducing compound monobenzone. We show monobenzone to increase melanocyte and melanoma cell immunogenicity by forming quinone-haptens to the tyrosinase protein and by inducing the release of tyrosinase- and melanoma antigen recognized by T cells-1 (MART-1)-containing CD63+ exosomes following melanosome oxidative stress induction. Monobenzone further augments the processing and shedding of melanocyte-differentiation antigens by inducing melanosome autophagy and enhanced tyrosinase ubiquitination, ultimately activating dendritic cells, which induced cytotoxic human melanoma-reactive T cells. These T cells effectively eradicate melanoma in vivo, as we have reported previously. Monobenzone thereby represents a promising and readily applicable compound for immunotherapy in melanoma patients.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Autofagia/efectos de los fármacos , Haptenos/metabolismo , Hidroquinonas/farmacología , Melanocitos/efectos de los fármacos , Melanosomas/efectos de los fármacos , Monofenol Monooxigenasa/metabolismo , Linfocitos T/efectos de los fármacos , Células Dendríticas/inmunología , Antígenos HLA-DR/análisis , Humanos , Lisosomas/metabolismo , Melaninas/biosíntesis , Melanocitos/inmunología , Melanoma/inmunología , Melanoma/terapia , Melanosomas/fisiología , Monofenol Monooxigenasa/inmunología , Especies Reactivas de Oxígeno/metabolismo , Pigmentación de la Piel/efectos de los fármacos , Linfocitos T/inmunología , Ubiquitinación
7.
PLoS One ; 5(5): e10626, 2010 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-20498710

RESUMEN

BACKGROUND: Presently melanoma still lacks adequate treatment options for metastatic disease. While melanoma is exceptionally challenging to standard regimens, it is suited for treatment with immunotherapy based on its immunogenicity. Since treatment-related skin depigmentation is considered a favourable prognostic sign during melanoma intervention, we here aimed at the reverse approach of directly inducing vitiligo as a shortcut to effective anti-melanoma immunity. METHODOLOGY AND PRINCIPAL FINDINGS: We developed an effective and simple to use form of immunotherapy by combining the topical skin-bleaching agent monobenzone with immune-stimulatory imiquimod cream and cytosine-guanine oligodeoxynucleotides (CpG) injections (MIC therapy). This powerful new approach promptly induced a melanoma antigen-specific immune response, which abolished subcutaneous B16.F10 melanoma growth in up to 85% of C57BL/6 mice. Importantly, this regimen induced over 100 days of tumor-free survival in up to 60% of the mice, and forcefully suppressed tumor growth upon re-challenge either 65- or 165 days after MIC treatment cessation. CONCLUSIONS: MIC therapy is effective in eradicating melanoma, by vigilantly incorporating NK-, B- and T cells in its therapeutic effect. Based on these results, the MIC regimen presents a high-yield, low-cost and simple therapy, readily applicable in the clinic.


Asunto(s)
Aminoquinolinas/uso terapéutico , Fosfatos de Dinucleósidos/uso terapéutico , Hidroquinonas/uso terapéutico , Inmunoterapia , Melanoma/tratamiento farmacológico , Pigmentación , Neoplasias Cutáneas/tratamiento farmacológico , Adyuvantes Inmunológicos/uso terapéutico , Aminoquinolinas/administración & dosificación , Aminoquinolinas/farmacología , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Proliferación Celular/efectos de los fármacos , Fosfatos de Dinucleósidos/administración & dosificación , Fosfatos de Dinucleósidos/farmacología , Hidroquinonas/administración & dosificación , Hidroquinonas/farmacología , Imiquimod , Inmunoglobulina G/inmunología , Inyecciones Subcutáneas , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Recuento de Leucocitos , Activación de Linfocitos/efectos de los fármacos , Depleción Linfocítica , Melanoma/inmunología , Ratones , Ratones Endogámicos C57BL , Pigmentación/efectos de los fármacos , Neoplasias Cutáneas/inmunología
8.
J Invest Dermatol ; 129(9): 2220-32, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19242513

RESUMEN

In vitiligo, cytotoxic T cells infiltrating the perilesional margin are suspected to be involved in the pathogenesis of the disease. However, it remains to be elucidated whether these T cells are a cause or a consequence of the depigmentation process. T cells we obtained from perilesional skin biopsies, were significantly enriched for melanocyte antigen recognition, compared with healthy skin-infiltrating T cells, and were reactive to melanocyte antigen-specific stimulation. Using a skin explant model, we were able to dissect the in situ activities of perilesional T cells in the effector phase of depigmentation. We show that these T cells could infiltrate autologous normally pigmented skin explants and efficiently kill melanocytes within this microenvironment. Interestingly, melanocyte apoptosis was accompanied by suprabasal keratinocyte apoptosis. Perilesional T cells did, however, not induce apoptosis in lesional skin, which is devoid of melanocytes, indicating the melanocyte-specific cytotoxic activity of these cells. Melanocyte killing correlated to local infiltration of perilesional T cells. Our data show that perilesional cytotoxic T cells eradicate pigment cells, the characteristic hallmark of vitiligo, thereby providing evidence of T cells being able to mediate targeted autoimmune tissue destruction.


Asunto(s)
Autoinmunidad , Melanocitos/patología , Piel/patología , Vitíligo/inmunología , Apoptosis , Citotoxicidad Inmunológica , Humanos , Interleucina-17/fisiología , Activación de Linfocitos , Melanocitos/inmunología , Piel/inmunología , Linfocitos T/inmunología , Vitíligo/etiología , Vitíligo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA