Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 576(7786): 293-300, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31802004

RESUMEN

Chimeric antigen receptor (CAR) T cells mediate anti-tumour effects in a small subset of patients with cancer1-3, but dysfunction due to T cell exhaustion is an important barrier to progress4-6. To investigate the biology of exhaustion in human T cells expressing CAR receptors, we used a model system with a tonically signaling CAR, which induces hallmark features of exhaustion6. Exhaustion was associated with a profound defect in the production of IL-2, along with increased chromatin accessibility of AP-1 transcription factor motifs and overexpression of the bZIP and IRF transcription factors that have been implicated in mediating dysfunction in exhausted T cells7-10. Here we show that CAR T cells engineered to overexpress the canonical AP-1 factor c-Jun have enhanced expansion potential, increased functional capacity, diminished terminal differentiation and improved anti-tumour potency in five different mouse tumour models in vivo. We conclude that a functional deficiency in c-Jun mediates dysfunction in exhausted human T cells, and that engineering CAR T cells to overexpress c-Jun renders them resistant to exhaustion, thereby addressing a major barrier to progress for this emerging class of therapeutic agents.


Asunto(s)
Proteínas Proto-Oncogénicas c-jun/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Epigénesis Genética , Regulación de la Expresión Génica , Humanos , Ratones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Receptores de Antígenos de Linfocitos T/genética , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/inmunología , Transcripción Genética
2.
Int J Cancer ; 143(7): 1706-1719, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29672841

RESUMEN

Familial aggregation is a significant risk factor for the development of thyroid cancer and familial non-medullary thyroid cancer (FNMTC) accounts for 5-7% of all NMTC. Whole exome sequencing analysis in the family affected by FNMTC with oncocytic features where our group previously identified a predisposing locus on chromosome 19p13.2, revealed a novel heterozygous mutation (c.400G > A, NM_012335; p.Gly134Ser) in exon 5 of MYO1F, mapping to the linkage locus. In the thyroid FRTL-5 cell model stably expressing the mutant MYO1F p.Gly134Ser protein, we observed an altered mitochondrial network, with increased mitochondrial mass and a significant increase in both intracellular and extracellular reactive oxygen species, compared to cells expressing the wild-type (wt) protein or carrying the empty vector. The mutation conferred a significant advantage in colony formation, invasion and anchorage-independent growth. These data were corroborated by in vivo studies in zebrafish, since we demonstrated that the mutant MYO1F p.Gly134Ser, when overexpressed, can induce proliferation in whole vertebrate embryos, compared to the wt one. MYO1F screening in additional 192 FNMTC families identified another variant in exon 7, which leads to exon skipping, and is predicted to alter the ATP-binding domain in MYO1F. Our study identified for the first time a role for MYO1F in NMTC.


Asunto(s)
Proliferación Celular , Embrión no Mamífero/patología , Mitocondrias/patología , Mutación , Miosina Tipo I/genética , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Apoptosis , Células Cultivadas , Niño , Cromosomas Humanos Par 19 , Embrión no Mamífero/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/genética , Mitocondrias/metabolismo , Miosina Tipo I/química , Miosina Tipo I/metabolismo , Consumo de Oxígeno , Linaje , Conformación Proteica , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/metabolismo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo , Adulto Joven , Pez Cebra
3.
Cancer Cell ; 42(1): 35-51.e8, 2024 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-38134936

RESUMEN

Chimeric antigen receptor T cells (CAR-Ts) have remarkable efficacy in liquid tumors, but limited responses in solid tumors. We conducted a Phase I trial (NCT02107963) of GD2 CAR-Ts (GD2-CAR.OX40.28.z.iC9), demonstrating feasibility and safety of administration in children and young adults with osteosarcoma and neuroblastoma. Since CAR-T efficacy requires adequate CAR-T expansion, patients were grouped into good or poor expanders across dose levels. Patient samples were evaluated by multi-dimensional proteomic, transcriptomic, and epigenetic analyses. T cell assessments identified naive T cells in pre-treatment apheresis associated with good expansion, and exhausted T cells in CAR-T products with poor expansion. Myeloid cell assessment identified CXCR3+ monocytes in pre-treatment apheresis associated with good expansion. Longitudinal analysis of post-treatment samples identified increased CXCR3- classical monocytes in all groups as CAR-T numbers waned. Together, our data uncover mediators of CAR-T biology and correlates of expansion that could be utilized to advance immunotherapies for solid tumor patients.


Asunto(s)
Neuroblastoma , Receptores Quiméricos de Antígenos , Niño , Adulto Joven , Humanos , Receptores Quiméricos de Antígenos/genética , Receptores de Antígenos de Linfocitos T/genética , Proteómica , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Linfocitos T , Neuroblastoma/patología , Tratamiento Basado en Trasplante de Células y Tejidos
4.
Cancer Cell ; 40(1): 53-69.e9, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-34971569

RESUMEN

Pediatric cancers often mimic fetal tissues and express proteins normally silenced postnatally that could serve as immune targets. We developed T cells expressing chimeric antigen receptors (CARs) targeting glypican-2 (GPC2), a fetal antigen expressed on neuroblastoma (NB) and several other solid tumors. CARs engineered using standard designs control NBs with transgenic GPC2 overexpression, but not those expressing clinically relevant GPC2 site density (∼5,000 molecules/cell, range 1-6 × 103). Iterative engineering of transmembrane (TM) and co-stimulatory domains plus overexpression of c-Jun lowered the GPC2-CAR antigen density threshold, enabling potent and durable eradication of NBs expressing clinically relevant GPC2 antigen density, without toxicity. These studies highlight the critical interplay between CAR design and antigen density threshold, demonstrate potent efficacy and safety of a lead GPC2-CAR candidate suitable for clinical testing, and credential oncofetal antigens as a promising class of targets for CAR T cell therapy of solid tumors.


Asunto(s)
Glipicanos/inmunología , Inmunoterapia Adoptiva , Neuroblastoma/tratamiento farmacológico , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Línea Celular Tumoral , Glipicanos/metabolismo , Humanos , Inmunoterapia/métodos , Neuroblastoma/patología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
5.
Sci Rep ; 11(1): 19138, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34580349

RESUMEN

Pleural mesothelioma is an aggressive malignancy with limited effective therapies. In order to identify therapeutic targets, we integrated SNP genotyping, sequencing and transcriptomics from tumours and low-passage patient-derived cells. Previously unrecognised deletions of SUFU locus (10q24.32), observed in 21% of 118 tumours, resulted in disordered expression of transcripts from Hedgehog pathways and the T-cell synapse including VISTA. Co-deletion of Interferon Type I genes and CDKN2A was present in half of tumours and was a predictor of poor survival. We also found previously unrecognised deletions in RB1 in 26% of cases and show sub-micromolar responses to downstream PLK1, CHEK1 and Aurora Kinase inhibitors in primary mesothelioma cells. Defects in Hippo pathways that included RASSF7 amplification and NF2 or LATS1/2 mutations were present in 50% of tumours and were accompanied by micromolar responses to the YAP1 inhibitor Verteporfin. Our results suggest new therapeutic avenues in mesothelioma and indicate targets and biomarkers for immunotherapy.


Asunto(s)
Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica/inmunología , Vía de Señalización Hippo/genética , Mesotelioma Maligno/genética , Neoplasias Pleurales/genética , Anciano , Anciano de 80 o más Años , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/antagonistas & inhibidores , Biopsia , Variaciones en el Número de Copia de ADN , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genómica , Vía de Señalización Hippo/efectos de los fármacos , Vía de Señalización Hippo/inmunología , Humanos , Masculino , Mesotelioma Maligno/tratamiento farmacológico , Mesotelioma Maligno/inmunología , Mesotelioma Maligno/patología , Persona de Mediana Edad , Mutación , Pleura/patología , Neoplasias Pleurales/tratamiento farmacológico , Neoplasias Pleurales/inmunología , Neoplasias Pleurales/patología , Cultivo Primario de Células , Secuenciación Completa del Genoma
6.
Science ; 372(6537)2021 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-33795428

RESUMEN

T cell exhaustion limits immune responses against cancer and is a major cause of resistance to chimeric antigen receptor (CAR)-T cell therapeutics. Using murine xenograft models and an in vitro model wherein tonic CAR signaling induces hallmark features of exhaustion, we tested the effect of transient cessation of receptor signaling, or rest, on the development and maintenance of exhaustion. Induction of rest through enforced down-regulation of the CAR protein using a drug-regulatable system or treatment with the multikinase inhibitor dasatinib resulted in the acquisition of a memory-like phenotype, global transcriptional and epigenetic reprogramming, and restored antitumor functionality in exhausted CAR-T cells. This work demonstrates that rest can enhance CAR-T cell efficacy by preventing or reversing exhaustion, and it challenges the notion that exhaustion is an epigenetically fixed state.


Asunto(s)
Dasatinib/farmacología , Epigénesis Genética , Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica , Regulación hacia Abajo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigenoma , Femenino , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Memoria Inmunológica , Activación de Linfocitos , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Masculino , Ratones , Neoplasias Experimentales/terapia , Dominios Proteicos , Estabilidad Proteica , Receptores Quiméricos de Antígenos/química , Receptores Quiméricos de Antígenos/inmunología , Transducción de Señal , Linfocitos T/metabolismo , Transcripción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Clin Cancer Res ; 25(16): 5156-5166, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31227497

RESUMEN

PURPOSE: Uveal melanoma is a primary malignancy of the eye with oncogenic mutations in GNAQ, GNA11, or CYSLTR2, and additional mutations in BAP1 (usually associated with LOH of Chr 3), SF3B1, or EIF1AX. There are other characteristic chromosomal alterations, but their significance is not clear. EXPERIMENTAL DESIGN: To investigate genes driving chromosomal alterations, we integrated copy number, transcriptome, and mutation data from three cohorts and followed up key findings. RESULTS: We observed significant enrichment of transcripts on chromosomes 1p, 3, 6, 8, and 16q and identified seven shared focal copy number alterations (FCNAs) on Chr 1p36, 2q37, 3, 6q25, 6q27, and 8q24. Integrated analyses revealed clusters of genes in focal copy number regions whose expression was associated with metastasis and worse overall survival. This included genes from Chr 1p36, 3p21, and 8q24.3. At Chr 6q27, we identified two tumors with homozygous deletion of PHF10/BAF45a and one with a frameshift mutation with concomitant loss of the wild-type allele. Downregulation of PHF10 in uveal melanoma cell lines and tumors altered a number of biological pathways including development and adhesion. These findings provide support for a role for PHF10 as a novel tumor suppressor at Chr 6q27. CONCLUSIONS: Integration of copy number, transcriptome, and mutation data revealed novel candidate genes playing a role in uveal melanoma pathogenesis and a potential tumor suppressor role for PHF10.


Asunto(s)
Transformación Celular Neoplásica/genética , Variaciones en el Número de Copia de ADN , Genes Supresores de Tumor , Proteínas de Homeodominio/genética , Melanoma/genética , Proteínas de Neoplasias/genética , Factores de Transcripción/genética , Neoplasias de la Úvea/genética , Metilación de ADN , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Homocigoto , Humanos , Melanoma/mortalidad , Melanoma/patología , Mutación , Polimorfismo de Nucleótido Simple , Pronóstico , Transcriptoma , Neoplasias de la Úvea/mortalidad , Neoplasias de la Úvea/patología
8.
Nat Commun ; 9(1): 116, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29317634

RESUMEN

Cancer is thought to arise through the accumulation of genomic aberrations evolving under Darwinian selection. However, it remains unclear when the aberrations associated with metastasis emerge during tumor evolution. Uveal melanoma (UM) is the most common primary eye cancer and frequently leads to metastatic death, which is strongly linked to BAP1 mutations. Accordingly, UM is ideally suited for studying the clonal evolution of metastatic competence. Here we analyze sequencing data from 151 primary UM samples using a customized bioinformatic pipeline, to improve detection of BAP1 mutations and infer the clonal relationships among genomic aberrations. Strikingly, we find BAP1 mutations and other canonical genomic aberrations usually arise in an early punctuated burst, followed by neutral evolution extending to the time of clinical detection. This implies that the metastatic proclivity of UM is "set in stone" early in tumor evolution and may explain why advances in primary treatment have not improved survival.


Asunto(s)
Melanoma/genética , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Neoplasias de la Úvea/genética , Análisis por Conglomerados , Variaciones en el Número de Copia de ADN , Metilación de ADN , Evolución Molecular , Humanos , Mutación , Secuenciación del Exoma
9.
J Invest Dermatol ; 137(5): e113-e118, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28411841

RESUMEN

The investigation of biological systems involving all organs of the body including the skin is in era of big data. This requires heavy-duty computational tools, and novel statistical methods. Microarrays have allowed the interrogation of thousands of common genetic markers in thousands of individuals from the same population (termed genome wide association studies or GWAS) to reveal common variation associated with disease or phenotype. These markers are usually single nucleotide polymorphisms (SNPs) that are relatively common in the population. In the case of dermatological diseases such as alopecia areata, vitiligo, psoriasis and atopic dermatitis, common variants have been identified that are associated with disease, and these provide insights into biological pathways and reveal possible novel drug targets. Other skin phenotypes such as acne, color and skin cancers are also being investigated with GWAS. Analyses of such large GWAS datasets require a consideration of a number of statistical issues including the testing of multiple markers, population substructure, and ultimately a requirement for replication. There are also issues regarding the missing heritability of disease that cannot be entirely explained with current GWAS approaches. Next generation sequencing technologies such as exome and genome sequencing of similar patient cohorts will reveal additional variants contributing to disease susceptibility. However, the data generated with these approaches will be orders of magnitude greater than that those generated with arrays, with concomitant challenges in the identification of disease causing variants.


Asunto(s)
Biología Computacional/métodos , Estudio de Asociación del Genoma Completo/métodos , Enfermedades de la Piel/genética , Predisposición Genética a la Enfermedad , Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Análisis por Micromatrices/métodos , Biología Molecular/métodos , Fenotipo , Polimorfismo de Nucleótido Simple , Enfermedades de la Piel/patología
11.
JAMA Ophthalmol ; 134(7): 728-33, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27123562

RESUMEN

IMPORTANCE: Frequent mutations have been described in the following 5 genes in uveal melanoma (UM): BAP1, EIF1AX, GNA11, GNAQ, and SF3B1. Understanding the prognostic significance of these mutations could facilitate their use in precision medicine. OBJECTIVE: To determine the associations between driver mutations, gene expression profile (GEP) classification, clinicopathologic features, and patient outcomes in UM. DESIGN, SETTING, AND PARTICIPANTS: Retrospective study of patients with UM treated by enucleation by a single ocular oncologist between November 1, 1998, and July 31, 2014. MAIN OUTCOMES AND MEASURES: Clinicopathologic features, patient outcomes, GEP classification (class 1 or class 2), and mutation status were recorded. RESULTS: The study cohort comprised 81 participants. Their mean age was 61.5 years, and 37% (30 of 81) were female. The GEP classification was class 1 in 35 of 81 (43%), class 2 in 42 of 81 (52%), and unknown in 4 of 81 (5%). BAP1 mutations were identified in 29 of 64 (45%), GNAQ mutations in 36 of 81 (44%), GNA11 mutations in 36 of 81 (44%), SF3B1 mutations in 19 of 81 (24%), and EIF1AX mutations in 14 of 81 (17%). Sixteen of the mutations in BAP1 and 6 of the mutations in EIF1AX were previously unreported in UM. GNAQ and GNA11 mutations were mutually exclusive. BAP1, SF3B1, and EIF1AX mutations were almost mutually exclusive with each other. Using multiple regression analysis, BAP1 mutations were associated with class 2 GEP and older patient. EIF1AX mutations were associated with class 1 GEP and the absence of ciliary body involvement. SF3B1 mutations were associated with younger patient age. GNAQ mutations were associated with the absence of ciliary body involvement and greater largest basal diameter. GNA11 mutations were not associated with any of the analyzed features. Using Cox proportional hazards modeling, class 2 GEP was the prognostic factor most strongly associated with metastasis (relative risk, 9.4; 95% CI, 3.1-28.5) and melanoma-specific mortality (relative risk, 15.7; 95% CI, 3.6-69.1) (P < .001 for both). After excluding GEP class, the presence of BAP1 mutations was the factor most strongly associated with metastasis (relative risk, 10.6; 95% CI, 3.4-33.5) and melanoma-specific mortality (relative risk, 9.0; 95% CI, 2.8-29.2) (P < .001 for both). CONCLUSIONS AND RELEVANCE: BAP1, SF3B1, and EIF1AX mutations occur during UM tumor progression in an almost mutually exclusive manner and are associated with different levels of metastatic risk. These mutations may have value as prognostic markers in UM.


Asunto(s)
Factor 1 Eucariótico de Iniciación/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Melanoma/genética , Mutación , Proteínas de Neoplasias/genética , Fosfoproteínas/genética , Factores de Empalme de ARN/genética , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Neoplasias de la Úvea/genética , Análisis Mutacional de ADN , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Melanoma/diagnóstico , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Pronóstico , Estudios Retrospectivos , Neoplasias de la Úvea/diagnóstico
12.
Nat Genet ; 45(2): 133-5, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23313955

RESUMEN

Uveal melanoma is the most common primary cancer of the eye and often results in fatal metastasis. Here, we describe mutations occurring exclusively at codon 625 of the SF3B1 gene, encoding splicing factor 3B subunit 1, in low-grade uveal melanomas with good prognosis. Thus, uveal melanoma is among a small group of cancers associated with SF3B1 mutations, and these mutations denote a distinct molecular subset of uveal melanomas.


Asunto(s)
Melanoma/genética , Fosfoproteínas/genética , Ribonucleoproteína Nuclear Pequeña U2/genética , Neoplasias de la Úvea/genética , Codón/genética , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN/genética , Humanos , Estimación de Kaplan-Meier , Análisis por Micromatrices , Mutación/genética , Fosfoproteínas/metabolismo , Factores de Empalme de ARN , Ribonucleoproteína Nuclear Pequeña U2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA