Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
3.
Immunooncol Technol ; 3: 1-7, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35757302

RESUMEN

Immune checkpoint blockade has significantly improved clinical outcomes for patients with non-small cell lung cancer (NSCLC) and other solid tumours, but many patients do not respond and acquired resistance is common. Aspects of the tumour microenvironment linked to clinical outcomes include the proportion of tumour-infiltrating lymphocytes (TIL), tumour programmed death ligand 1 ( PD-L1) score and tumour mutation burden. Adoptive cell therapy (ACT), a technique that works by infusing ex vivo expanded T lymphocytes to increase the effector cell pool in tumours, is anticipated to become a viable therapeutic option for patients with solid tumours, akin to chimeric antigen receptor T cell (CAR-T) therapy in haematological malignancies. TIL therapy has shown durable clinical responses in heavily pre-treated patients with melanoma and other solid tumours. We review the experience of ACT with TILs and the recent evidence that clonal neoantigens might be the most relevant immunotherapeutic targets in heterogeneous solid tumours such as NSCLC. Clonal (or truncal) neoantigens arise from the earliest mutagenic events in tumour evolution, and are retained over time in all tumour cells within a patient, making them the ideal target for T cell therapy. NSCLC has one of the highest clonal mutation burdens of all cancers through exposure to carcinogens in tobacco smoke, providing a strong rationale to develop clonal neoantigen reactive T cells (cNeT) for this indication. The first treatment modality to test this concept clinically is ATL001, a cNeT product that is derived from autologous TILs and enriched for T cells specifically recognizing clonal neoantigenic epitopes by selective expansion. Clinical studies of ATL001 will commence in 2019.

4.
Cell ; 173(3): 595-610.e11, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29656894

RESUMEN

The evolutionary features of clear-cell renal cell carcinoma (ccRCC) have not been systematically studied to date. We analyzed 1,206 primary tumor regions from 101 patients recruited into the multi-center prospective study, TRACERx Renal. We observe up to 30 driver events per tumor and show that subclonal diversification is associated with known prognostic parameters. By resolving the patterns of driver event ordering, co-occurrence, and mutual exclusivity at clone level, we show the deterministic nature of clonal evolution. ccRCC can be grouped into seven evolutionary subtypes, ranging from tumors characterized by early fixation of multiple mutational and copy number drivers and rapid metastases to highly branched tumors with >10 subclonal drivers and extensive parallel evolution associated with attenuated progression. We identify genetic diversity and chromosomal complexity as determinants of patient outcome. Our insights reconcile the variable clinical behavior of ccRCC and suggest evolutionary potential as a biomarker for both intervention and surveillance.


Asunto(s)
Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Neoplasias Renales/genética , Neoplasias Renales/patología , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Biomarcadores de Tumor , Cromosomas , Evolución Clonal , Progresión de la Enfermedad , Evolución Molecular , Femenino , Heterogeneidad Genética , Variación Genética , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Modelos Estadísticos , Mutación , Metástasis de la Neoplasia , Fenotipo , Filogenia , Pronóstico , Estudios Prospectivos , Análisis de Secuencia de ADN
5.
N Engl J Med ; 376(22): 2109-2121, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28445112

RESUMEN

BACKGROUND: Among patients with non-small-cell lung cancer (NSCLC), data on intratumor heterogeneity and cancer genome evolution have been limited to small retrospective cohorts. We wanted to prospectively investigate intratumor heterogeneity in relation to clinical outcome and to determine the clonal nature of driver events and evolutionary processes in early-stage NSCLC. METHODS: In this prospective cohort study, we performed multiregion whole-exome sequencing on 100 early-stage NSCLC tumors that had been resected before systemic therapy. We sequenced and analyzed 327 tumor regions to define evolutionary histories, obtain a census of clonal and subclonal events, and assess the relationship between intratumor heterogeneity and recurrence-free survival. RESULTS: We observed widespread intratumor heterogeneity for both somatic copy-number alterations and mutations. Driver mutations in EGFR, MET, BRAF, and TP53 were almost always clonal. However, heterogeneous driver alterations that occurred later in evolution were found in more than 75% of the tumors and were common in PIK3CA and NF1 and in genes that are involved in chromatin modification and DNA damage response and repair. Genome doubling and ongoing dynamic chromosomal instability were associated with intratumor heterogeneity and resulted in parallel evolution of driver somatic copy-number alterations, including amplifications in CDK4, FOXA1, and BCL11A. Elevated copy-number heterogeneity was associated with an increased risk of recurrence or death (hazard ratio, 4.9; P=4.4×10-4), which remained significant in multivariate analysis. CONCLUSIONS: Intratumor heterogeneity mediated through chromosome instability was associated with an increased risk of recurrence or death, a finding that supports the potential value of chromosome instability as a prognostic predictor. (Funded by Cancer Research UK and others; TRACERx ClinicalTrials.gov number, NCT01888601 .).


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Inestabilidad Cromosómica , Heterogeneidad Genética , Neoplasias Pulmonares/genética , Mutación , Recurrencia Local de Neoplasia/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Variaciones en el Número de Copia de ADN , Supervivencia sin Enfermedad , Evolución Molecular , Exoma , Femenino , Humanos , Neoplasias Pulmonares/mortalidad , Masculino , Filogenia , Pronóstico , Estudios Prospectivos , Factores de Riesgo , Análisis de Secuencia de ADN/métodos
6.
Cancer Discov ; 5(8): 821-831, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26003801

RESUMEN

UNLABELLED: Esophageal adenocarcinomas are associated with a dismal prognosis. Deciphering the evolutionary history of this disease may shed light on therapeutically tractable targets and reveal dynamic mutational processes during the disease course and following neoadjuvant chemotherapy (NAC). We exome sequenced 40 tumor regions from 8 patients with operable esophageal adenocarcinomas, before and after platinum-containing NAC. This revealed the evolutionary genomic landscape of esophageal adenocarcinomas with the presence of heterogeneous driver mutations, parallel evolution, early genome-doubling events, and an association between high intratumor heterogeneity and poor response to NAC. Multiregion sequencing demonstrated a significant reduction in thymine to guanine mutations within a CpTpT context when comparing early and late mutational processes and the presence of a platinum signature with enrichment of cytosine to adenine mutations within a CpC context following NAC. Esophageal adenocarcinomas are characterized by early chromosomal instability leading to amplifications containing targetable oncogenes persisting through chemotherapy, providing a rationale for future therapeutic approaches. SIGNIFICANCE: This work illustrates dynamic mutational processes occurring during esophageal adenocarcinoma evolution and following selective pressures of platinum exposure, emphasizing the iatrogenic impact of therapy on cancer evolution. Identification of amplifications encoding targetable oncogenes maintained through NAC suggests the presence of stable vulnerabilities, unimpeded by cytotoxics, suitable for therapeutic intervention.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/genética , Evolución Molecular , Genoma Humano , Adenocarcinoma/patología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Evolución Clonal , Biología Computacional , Variaciones en el Número de Copia de ADN , Neoplasias Esofágicas/patología , Exoma , Amplificación de Genes , Variación Genética , Inestabilidad Genómica , Genómica/métodos , Humanos , Mutación , Terapia Neoadyuvante , Platino (Metal)/administración & dosificación
7.
Nat Commun ; 6: 6336, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25790038

RESUMEN

Papillary renal cell carcinoma (pRCC) is an important subtype of kidney cancer with a problematic pathological classification and highly variable clinical behaviour. Here we sequence the genomes or exomes of 31 pRCCs, and in four tumours, multi-region sequencing is undertaken. We identify BAP1, SETD2, ARID2 and Nrf2 pathway genes (KEAP1, NHE2L2 and CUL3) as probable drivers, together with at least eight other possible drivers. However, only ~10% of tumours harbour detectable pathogenic changes in any one driver gene, and where present, the mutations are often predicted to be present within cancer sub-clones. We specifically detect parallel evolution of multiple SETD2 mutations within different sub-regions of the same tumour. By contrast, large copy number gains of chromosomes 7, 12, 16 and 17 are usually early, monoclonal changes in pRCC evolution. The predominance of large copy number variants as the major drivers for pRCC highlights an unusual mode of tumorigenesis that may challenge precision medicine approaches.


Asunto(s)
Carcinoma de Células Renales/genética , Cromosomas/ultraestructura , Neoplasias Renales/genética , Mutación , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/química , Mapeo Cromosómico , Variaciones en el Número de Copia de ADN , Exoma , Exones , Femenino , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Pérdida de Heterocigocidad , Masculino , Persona de Mediana Edad , Filogenia , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN
8.
Science ; 346(6206): 251-6, 2014 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-25301630

RESUMEN

Spatial and temporal dissection of the genomic changes occurring during the evolution of human non-small cell lung cancer (NSCLC) may help elucidate the basis for its dismal prognosis. We sequenced 25 spatially distinct regions from seven operable NSCLCs and found evidence of branched evolution, with driver mutations arising before and after subclonal diversification. There was pronounced intratumor heterogeneity in copy number alterations, translocations, and mutations associated with APOBEC cytidine deaminase activity. Despite maintained carcinogen exposure, tumors from smokers showed a relative decrease in smoking-related mutations over time, accompanied by an increase in APOBEC-associated mutations. In tumors from former smokers, genome-doubling occurred within a smoking-signature context before subclonal diversification, which suggested that a long period of tumor latency had preceded clinical detection. The regionally separated driver mutations, coupled with the relentless and heterogeneous nature of the genome instability processes, are likely to confound treatment success in NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/genética , Heterogeneidad Genética , Inestabilidad Genómica , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Desaminasas APOBEC-1 , Carcinógenos/toxicidad , Carcinoma de Pulmón de Células no Pequeñas/inducido químicamente , Citidina Desaminasa/genética , Evolución Molecular , Dosificación de Gen , Humanos , Neoplasias Pulmonares/inducido químicamente , Mutación , Recurrencia Local de Neoplasia/genética , Pronóstico , Fumar/efectos adversos , Translocación Genética , Células Tumorales Cultivadas
9.
Genome Biol ; 15(9): 470, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25270265

RESUMEN

Intra-tumor heterogeneity concerns the existence of genetically different subclones within the same tumor. Single sample quantification of heterogeneity relies on precise determination of chromosomal copy numbers throughout the genome, and an assessment of whether identified mutation variant allele fractions match clonal or subclonal copy numbers. We discuss these issues using data from SNP arrays, whole exome sequencing and pathologist purity estimates on several breast cancers characterized by ERBB2 amplification. We show that chromosomal copy numbers can only be estimated from SNP array signals or sequencing depths for subclonal tumor samples with simple subclonal architectures under certain assumptions.


Asunto(s)
Aneuploidia , Neoplasias de la Mama/genética , Polimorfismo de Nucleótido Simple , Femenino , Dosificación de Gen , Frecuencia de los Genes , Heterogeneidad Genética , Humanos , Modelos Genéticos , Modelos Estadísticos , Anotación de Secuencia Molecular , Análisis de Secuencia de ADN
10.
PLoS Biol ; 12(7): e1001906, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25003521

RESUMEN

The importance of intratumour genetic and functional heterogeneity is increasingly recognised as a driver of cancer progression and survival outcome. Understanding how tumour clonal heterogeneity impacts upon therapeutic outcome, however, is still an area of unmet clinical and scientific need. TRACERx (TRAcking non-small cell lung Cancer Evolution through therapy [Rx]), a prospective study of patients with primary non-small cell lung cancer (NSCLC), aims to define the evolutionary trajectories of lung cancer in both space and time through multiregion and longitudinal tumour sampling and genetic analysis. By following cancers from diagnosis to relapse, tracking the evolutionary trajectories of tumours in relation to therapeutic interventions, and determining the impact of clonal heterogeneity on clinical outcomes, TRACERx may help to identify novel therapeutic targets for NSCLC and may also serve as a model applicable to other cancer types.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Progresión de la Enfermedad , Neoplasias Pulmonares/genética , Antígenos de Neoplasias , Biomarcadores de Tumor/análisis , Resistencia a Antineoplásicos , Humanos , Estudios Longitudinales , Metástasis de la Neoplasia , Resultado del Tratamiento
11.
Nat Genet ; 46(3): 225-233, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24487277

RESUMEN

Clear cell renal carcinomas (ccRCCs) can display intratumor heterogeneity (ITH). We applied multiregion exome sequencing (M-seq) to resolve the genetic architecture and evolutionary histories of ten ccRCCs. Ultra-deep sequencing identified ITH in all cases. We found that 73-75% of identified ccRCC driver aberrations were subclonal, confounding estimates of driver mutation prevalence. ITH increased with the number of biopsies analyzed, without evidence of saturation in most tumors. Chromosome 3p loss and VHL aberrations were the only ubiquitous events. The proportion of C>T transitions at CpG sites increased during tumor progression. M-seq permits the temporal resolution of ccRCC evolution and refines mutational signatures occurring during tumor development.


Asunto(s)
Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Mutación , Fosfatidilinositol 3-Quinasa Clase I , Islas de CpG , Variaciones en el Número de Copia de ADN , Proteínas de Unión al ADN , Progresión de la Enfermedad , Evolución Molecular , Exoma , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Proteínas Nucleares/genética , Fosfatidilinositol 3-Quinasas/genética , Filogenia , Polimorfismo de Nucleótido Simple , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
12.
J Lipid Res ; 54(12): 3491-505, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24103848

RESUMEN

The purpose of this study was to determine the core biological processes perturbed in the subcutaneous adipose tissue of familial combined hyperlipidemia (FCHL) patients. Annotation of FCHL and control microarray datasets revealed a distinctive FCHL transcriptome, characterized by gene expression changes regulating five overlapping systems: the cytoskeleton, cell adhesion and extracellular matrix; vesicular trafficking; lipid homeostasis; and cell cycle and apoptosis. Expression values for the cell-cycle inhibitor CDKN2B were increased, replicating data from an independent FCHL cohort. In 3T3-L1 cells, CDKN2B knockdown induced C/EBPα expression and lipid accumulation. The minor allele at SNP site rs1063192 (C) was predicted to create a perfect seed for the human miRNA-323b-5p. A miR-323b-5p mimic significantly reduced endogenous CDKN2B protein levels and the activity of a CDKN2B 3'UTR luciferase reporter carrying the rs1063192 C allele. Although the allele displayed suggestive evidence of association with reduced CDKN2B mRNA in the MuTHER adipose tissue dataset, family studies suggest the association between increased CDKN2B expression and FCHL-lipid abnormalities is driven by factors external to this gene locus. In conclusion, from a comparative annotation analysis of two separate FCHL adipose tissue transcriptomes and a subsequent focus on CDKN2B, we propose that dysfunctional adipogenesis forms an integral part of FCHL pathogenesis.


Asunto(s)
Tejido Adiposo/metabolismo , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Regulación de la Expresión Génica , Hiperlipidemia Familiar Combinada/genética , Células 3T3-L1 , Adipogénesis/genética , Tejido Adiposo/patología , Animales , Ciclo Celular/genética , Células HEK293 , Haplotipos , Humanos , Hiperlipidemia Familiar Combinada/patología , Masculino , Ratones , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA