Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Hum Mol Genet ; 27(23): 4145-4156, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30060076

RESUMEN

Melanoma heritability is among the highest for cancer and single nucleotide polymorphisms (SNPs) contribute to it. To date, only SNPs that reached statistical significance in genome-wide association studies or few candidate SNPs have been included in melanoma risk prediction models. We compared four approaches for building polygenic risk scores (PRS) using 12 874 melanoma cases and 23 203 controls from Melanoma Meta-Analysis Consortium as a training set, and newly genotyped 3102 cases and 2301 controls from the MelaNostrum consortium for validation. We estimated adjusted odds ratios (ORs) for melanoma risk using traditional melanoma risk factors and the PRS with the largest area under the receiver operator characteristics curve (AUC). We estimated absolute risks combining the PRS and other risk factors, with age- and sex-specific melanoma incidence and competing mortality rates from Italy as an example. The best PRS, including 204 SNPs (AUC = 64.4%; 95% confidence interval (CI) = 63-65.8%), developed using winner's curse estimate corrections, had a per-quintile OR = 1.35 (95% CI = 1.30-1.41), corresponding to a 3.33-fold increase comparing the 5th to the 1st PRS quintile. The AUC improvement by adding the PRS was up to 7%, depending on adjusted factors and country. The 20-year absolute risk estimates based on the PRS, nevus count and pigmentation characteristics for a 60-year-old Italian man ranged from 0.5 to 11.8% (relative risk  = 26.34), indicating good separation.


Asunto(s)
Predisposición Genética a la Enfermedad , Melanoma/genética , Nevo/genética , Neoplasias Cutáneas/genética , Adulto , Anciano , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Italia , Masculino , Melanoma/epidemiología , Melanoma/patología , Persona de Mediana Edad , Herencia Multifactorial/genética , Nevo/epidemiología , Nevo/patología , Polimorfismo de Nucleótido Simple , Medición de Riesgo , Factores de Riesgo , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/patología , Melanoma Cutáneo Maligno
2.
Int J Cancer ; 135(7): 1625-33, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24535833

RESUMEN

We report the association of an inherited variant located upstream of the poly(adenosine diphosphate-ribose) polymerase 1 (PARP1) gene (rs2249844), with survival in 11 BioGenoMEL melanoma cohorts. The gene encodes a protein involved in a number of cellular processes including single-strand DNA repair. Survival analysis was conducted for each cohort using proportional hazards regression adjusting for factors known to be associated with survival. Survival was measured as overall survival (OS) and, where available, melanoma-specific survival (MSS). Results were combined using random effects meta-analysis. Evidence for a role of the PARP1 protein in melanoma ulceration and survival was investigated by testing gene expression levels taken from formalin-fixed paraffin-embedded tumors. A significant association was seen for inheritance of the rarer variant of PARP1, rs2249844 with OS (hazard ratio (HR) = 1.16 per allele, 95% confidence interval (CI) 1.04-1.28, p = 0.005, eleven cohorts) and MSS (HR = 1.20 per allele, 95% CI 1.01-1.39, p = 0.03, eight cohorts). We report bioinformatic data supportive of a functional effect for rs2249844. Higher levels of PARP1 gene expression in tumors were shown to be associated with tumor ulceration and poorer OS.


Asunto(s)
Predisposición Genética a la Enfermedad , Melanoma/genética , Melanoma/mortalidad , Poli(ADP-Ribosa) Polimerasas/genética , Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo , ADN de Neoplasias/genética , Estudios de Seguimiento , Humanos , Poli(ADP-Ribosa) Polimerasa-1 , Reacción en Cadena de la Polimerasa , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia
3.
Nat Commun ; 11(1): 597, 2020 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-32001714

RESUMEN

Physical activity has been associated with lower risks of breast and colorectal cancer in epidemiological studies; however, it is unknown if these associations are causal or confounded. In two-sample Mendelian randomisation analyses, using summary genetic data from the UK Biobank and GWA consortia, we found that a one standard deviation increment in average acceleration was associated with lower risks of breast cancer (odds ratio [OR]: 0.51, 95% confidence interval [CI]: 0.27 to 0.98, P-value = 0.04) and colorectal cancer (OR: 0.66, 95% CI: 0.48 to 0.90, P-value = 0.01). We found similar magnitude inverse associations for estrogen positive (ER+ve) breast cancer and for colon cancer. Our results support a potentially causal relationship between higher physical activity levels and lower risks of breast cancer and colorectal cancer. Based on these data, the promotion of physical activity is probably an effective strategy in the primary prevention of these commonly diagnosed cancers.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias Colorrectales/genética , Ejercicio Físico , Predisposición Genética a la Enfermedad , Análisis de la Aleatorización Mendeliana , Acelerometría , Femenino , Humanos , Oportunidad Relativa , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo
6.
J Natl Cancer Inst ; 109(12)2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29522175

RESUMEN

Background: Extraordinary progress has been made in our understanding of common variants in many diseases, including melanoma. Because the contribution of rare coding variants is not as well characterized, we performed an exome-wide, gene-based association study of familial cutaneous melanoma (CM) and ocular melanoma (OM). Methods: Using 11 990 jointly processed individual DNA samples, whole-exome sequencing was performed, followed by large-scale joint variant calling using GATK (Genome Analysis ToolKit). PLINK/SEQ was used for statistical analysis of genetic variation. Four models were used to estimate the association among different types of variants. In vitro functional validation was performed using three human melanoma cell lines in 2D and 3D proliferation assays. In vivo tumor growth was assessed using xenografts of human melanoma A375 melanoma cells in nude mice (eight mice per group). All statistical tests were two-sided. Results: Strong signals were detected for CDKN2A (Pmin = 6.16 × 10-8) in the CM cohort (n = 273) and BAP1 (Pmin = 3.83 × 10-6) in the OM (n = 99) cohort. Eleven genes that exhibited borderline association (P < 10-4) were independently validated using The Cancer Genome Atlas melanoma cohort (379 CM, 47 OM) and a matched set of 3563 European controls with CDKN2A (P = .009), BAP1 (P = .03), and EBF3 (P = 4.75 × 10-4), a candidate risk locus, all showing evidence of replication. EBF3 was then evaluated using germline data from a set of 132 familial melanoma cases and 4769 controls of UK origin (joint P = 1.37 × 10-5). Somatically, loss of EBF3 expression correlated with progression, poorer outcome, and high MITF tumors. Functionally, induction of EBF3 in melanoma cells reduced cell growth in vitro, retarded tumor formation in vivo, and reduced MITF levels. Conclusions: The results of this large rare variant germline association study further define the mutational landscape of hereditary melanoma and implicate EBF3 as a possible CM predisposition gene.


Asunto(s)
Biomarcadores de Tumor/genética , Secuenciación del Exoma/métodos , Neoplasias del Ojo/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Melanoma/genética , Neoplasias Cutáneas/genética , Estudios de Casos y Controles , Exoma , Neoplasias del Ojo/patología , Mutación de Línea Germinal , Humanos , Melanoma/patología , Pronóstico , Neoplasias Cutáneas/patología , Tasa de Supervivencia , Factores de Transcripción
7.
Nat Commun ; 7: 12064, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27403562

RESUMEN

The major genetic determinants of cutaneous melanoma risk in the general population are disruptive variants (R alleles) in the melanocortin 1 receptor (MC1R) gene. These alleles are also linked to red hair, freckling, and sun sensitivity, all of which are known melanoma phenotypic risk factors. Here we report that in melanomas and for somatic C>T mutations, a signature linked to sun exposure, the expected single-nucleotide variant count associated with the presence of an R allele is estimated to be 42% (95% CI, 15-76%) higher than that among persons without an R allele. This figure is comparable to the expected mutational burden associated with an additional 21 years of age. We also find significant and similar enrichment of non-C>T mutation classes supporting a role for additional mutagenic processes in melanoma development in individuals carrying R alleles.


Asunto(s)
Mutación de Línea Germinal/genética , Melanoma/genética , Acumulación de Mutaciones , Receptor de Melanocortina Tipo 1/genética , Neoplasias Cutáneas/genética , Anciano , Alelos , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad , Variación Genética , Color del Cabello , Neoplasias de Cabeza y Cuello/genética , Humanos , Masculino , Melanoma/patología , Melanosis , Persona de Mediana Edad , Mutación , Invasividad Neoplásica , Polimorfismo de Nucleótido Simple , Neoplasias Cutáneas/patología , Pigmentación de la Piel
9.
Genes Chromosomes Cancer ; 47(1): 34-42, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17943970

RESUMEN

Mutations in the KIT gene occur in approximately 8% of all testicular germ cell tumors (TGCT) and KIT is the most frequently mutated known cancer gene. One report has shown that 93% of patients with bilateral disease have a mutation at codon 816 of the KIT gene. Importantly, this suggests that the identification of a mutation in KIT is predictive of the development of a contralateral TGCT. We investigated the frequency and type of mutations in KIT in a series of 220 tumors from 211 patients with TGCTs and extragonadal germ cell tumors. In 170 patients with unilateral TGCT and no additional germ cell tumour, we identified one exon 11 mutation in a patient with unilateral TGCT and eight activating KIT mutations in exon 17 (9/175, 5.1%). In 32 patients with bilateral TGCT, one patient had an activating KIT mutation in exon 17 (3.1%). The incidence of activating KIT mutations in sporadic TGCT vs. familial TGCT was not significantly different. All mutations were identified in seminomas. Three extragonadal primary germ cell tumors were examined and in one tumor an activating KIT mutation was demonstrated in the pineal germinoma. Interestingly, this mutation was also seen in the patient's testicular seminoma. We find no evidence for an increased frequency of KIT mutations in bilateral TGCT.


Asunto(s)
Mutación , Proteínas Proto-Oncogénicas c-kit/genética , Seminoma/genética , Neoplasias Testiculares/genética , Adulto , Sustitución de Aminoácidos/genética , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Seminoma/patología , Neoplasias Testiculares/patología
10.
J Natl Cancer Inst ; 100(11): 784-95, 2008 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-18505964

RESUMEN

BACKGROUND: The tumor suppressors p14(ARF) (ARF) and p16(INK4A) (p16) are encoded by overlapping reading frames at the CDKN2A/INK4A locus on chromosome 9p21. In human melanoma, the accumulated evidence has suggested that the predominant tumor suppressor at 9p21 is p16, not ARF. However, recent observations from melanoma-prone families and murine melanoma models suggest a p16-independent tumor suppressor role for ARF. We analyzed a group of melanoma metastases and cell lines to investigate directly whether somatic alterations to the ARF gene support its role as a p16-independent tumor suppressor in human melanoma, assuming that two alterations (genetic and/or epigenetic) would be required to inactivate a gene. METHODS: We examined the p16/ARF locus in 60 melanoma metastases from 58 patients and in 9 human melanoma cell lines using multiplex ligation-dependent probe amplification and multiplex polymerase chain reaction (PCR) to detect deletions, methylation-specific PCR to detect promoter methylation, direct sequencing to detect mutations affecting ARF and p16, and, in a subset of 20 tumors, immunohistochemistry to determine the effect of these alterations on p16 protein expression. All statistical tests were two-sided. RESULTS: We observed two or more alterations to the ARF gene in 26/60 (43%) metastases. The p16 gene sustained two or more alterations in 13/60 (22%) metastases (P = .03). Inactivation of ARF in the presence of wild-type p16 was seen in 18/60 (30%) metastases. CONCLUSION: Genetic and epigenetic analyses of the human 9p21 locus indicate that modifications of ARF occur independently of p16 inactivation in human melanoma and suggest that ARF is more frequently inactivated than p16.


Asunto(s)
Silenciador del Gen , Genes p16 , Melanoma/genética , Melanoma/patología , Proteína p14ARF Supresora de Tumor/genética , Animales , Línea Celular Tumoral , Cromosomas Humanos Par 9 , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Metilación de ADN , Eliminación de Gen , Humanos , Immunoblotting , Inmunohistoquímica , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones , Repeticiones de Microsatélite , Mutación , Proteínas de Neoplasias/genética , Reacción en Cadena de la Polimerasa/métodos , Regiones Promotoras Genéticas
11.
Pharmacogenet Genomics ; 15(10): 713-21, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16141797

RESUMEN

OBJECTIVES: Regular continuous non-steroidal anti-inflammatory drug (NSAID) use has been associated with a reduction in risk of colorectal cancer. Our objective was to investigate whether or not a number of the polymorphic genes involved in the metabolism of NSAIDs, including cytochrome P450 s (CYPs), act as modifiers of this protective effect. METHODS: As part of a multi-centre case-control study, 478 colorectal cancer patients and 733 controls (433 matched case-control pairs) answered questions on NSAID use. These individuals were then genotyped for common polymorphisms in P450 CYP2C8, P450 CYP2C9, UDP-glucuronosyl transferase (UGT)1A6 and peroxisome proliferator-activated receptor isoforms delta and gamma (PPARdelta and PPARgamma). RESULTS AND CONCLUSION: Our study confirmed the reduction in risk of colorectal cancer with regular NSAID use (odds ratio (OR) = 0.73, 95% confidence interval (CI) (0.56, 0.95)) but showed that none of the polymorphic genes studied appeared to modify the protective effect of regular NSAID use.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Biomarcadores de Tumor/genética , Neoplasias del Colon/genética , Proteínas de Neoplasias/genética , PPAR delta/genética , Polimorfismo Genético , Sustancias Protectoras/uso terapéutico , Anciano , Anciano de 80 o más Años , Hidrocarburo de Aril Hidroxilasas/genética , Estudios de Casos y Controles , Neoplasias del Colon/prevención & control , Citocromo P-450 CYP2C8 , Citocromo P-450 CYP2C9 , Predisposición Genética a la Enfermedad , Glucuronosiltransferasa/genética , Humanos , Persona de Mediana Edad , PPAR gamma/genética , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA