Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nat Commun ; 12(1): 117, 2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33402692

RESUMEN

Nasopharyngeal cancer (NPC), endemic in Southeast Asia, lacks effective diagnostic and therapeutic strategies. Even in high-income countries the 5-year survival rate for stage IV NPC is less than 40%. Here we report high somatostatin receptor 2 (SSTR2) expression in multiple clinical cohorts comprising 402 primary, locally recurrent and metastatic NPCs. We show that SSTR2 expression is induced by the Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) via the NF-κB pathway. Using cell-based and preclinical rodent models, we demonstrate the therapeutic potential of SSTR2 targeting using a cytotoxic drug conjugate, PEN-221, which is found to be superior to FDA-approved SSTR2-binding cytostatic agents. Furthermore, we reveal significant correlation of SSTR expression with increased rates of survival and report in vivo uptake of the SSTR2-binding 68Ga-DOTA-peptide radioconjugate in PET-CT scanning in a clinical trial of NPC patients (NCT03670342). These findings reveal a key role in EBV-associated NPC for SSTR2 in infection, imaging, targeted therapy and survival.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Regulación Neoplásica de la Expresión Génica , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas , Recurrencia Local de Neoplasia , Receptores de Somatostatina , Proteínas de la Matriz Viral , Animales , Femenino , Humanos , Masculino , Ratones , Antineoplásicos/farmacología , Línea Celular Tumoral , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/mortalidad , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/efectos de los fármacos , Herpesvirus Humano 4/crecimiento & desarrollo , Herpesvirus Humano 4/patogenicidad , Interacciones Huésped-Patógeno/genética , Metástasis Linfática , Ratones Desnudos , Terapia Molecular Dirigida , Carcinoma Nasofaríngeo/tratamiento farmacológico , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/mortalidad , Carcinoma Nasofaríngeo/virología , Neoplasias Nasofaríngeas/tratamiento farmacológico , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/mortalidad , Neoplasias Nasofaríngeas/virología , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/virología , FN-kappa B/genética , FN-kappa B/metabolismo , Octreótido/farmacología , Tomografía Computarizada por Tomografía de Emisión de Positrones , Receptores de Somatostatina/antagonistas & inhibidores , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Transducción de Señal , Análisis de Supervivencia , Proteínas de la Matriz Viral/antagonistas & inhibidores , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Commun Biol ; 3(1): 740, 2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33288854

RESUMEN

Recent data suggest that Pancreatic Neuroendocrine Tumours (PanNETs) originate from α- or ß-cells of the islets of Langerhans. The majority of PanNETs are non-functional and do not express cell-type specific hormones. In the current study we examine whether tumour DNA methylation (DNAme) profiling combined with genomic data is able to identify cell of origin and to reveal pathways involved in PanNET progression. We analyse genome-wide DNAme data of 125 PanNETs and sorted α- and ß-cells. To confirm cell identity, we investigate ARX and PDX1 expression. Based on epigenetic similarities, PanNETs cluster in α-like, ß-like and intermediate tumours. The epigenetic similarity to α-cells progressively decreases in the intermediate tumours, which present unclear differentiation. Specific transcription factor methylation and expression vary in the respective α/ß-tumour groups. Depending on DNAme similarity to α/ß-cells, PanNETs have different mutational spectra, stage of the disease and prognosis, indicating potential means of PanNET progression.


Asunto(s)
Epigénesis Genética , Regulación Neoplásica de la Expresión Génica/fisiología , Tumores Neuroendocrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Variaciones en el Número de Copia de ADN , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética
3.
Cancer Discov ; 10(10): 1489-1499, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32690541

RESUMEN

Before squamous cell lung cancer develops, precancerous lesions can be found in the airways. From longitudinal monitoring, we know that only half of such lesions become cancer, whereas a third spontaneously regress. Although recent studies have described the presence of an active immune response in high-grade lesions, the mechanisms underpinning clinical regression of precancerous lesions remain unknown. Here, we show that host immune surveillance is strongly implicated in lesion regression. Using bronchoscopic biopsies from human subjects, we find that regressive carcinoma in situ lesions harbor more infiltrating immune cells than those that progress to cancer. Moreover, molecular profiling of these lesions identifies potential immune escape mechanisms specifically in those that progress to cancer: antigen presentation is impaired by genomic and epigenetic changes, CCL27-CCR10 signaling is upregulated, and the immunomodulator TNFSF9 is downregulated. Changes appear intrinsic to the carcinoma in situ lesions, as the adjacent stroma of progressive and regressive lesions are transcriptomically similar. SIGNIFICANCE: Immune evasion is a hallmark of cancer. For the first time, this study identifies mechanisms by which precancerous lesions evade immune detection during the earliest stages of carcinogenesis and forms a basis for new therapeutic strategies that treat or prevent early-stage lung cancer.See related commentary by Krysan et al., p. 1442.This article is highlighted in the In This Issue feature, p. 1426.


Asunto(s)
Carcinoma de Células Escamosas/inmunología , Vigilancia Inmunológica/inmunología , Neoplasias Pulmonares/inmunología , Humanos
4.
Endocr Relat Cancer ; 26(9): R519-R544, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31252410

RESUMEN

Neuroendocrine neoplasms (NENs) are a relatively rare group of heterogeneous tumours originating from neuroendocrine cells found throughout the body. Pancreatic NENs (PanNENs) are the second most common pancreatic malignancy accounting for 1-3% of all neoplasms developing in the pancreas. Despite having a low background mutation rate, driver mutations in MEN1, DAXX/ATRX and mTOR pathway genes (PTEN, TSC1/2) are implicated in disease development and progression. Their increased incidence coupled with advances in sequencing technologies has reignited the interest in PanNEN research and has accelerated the acquisition of molecular data. Studies utilising such technological advances have further enriched our knowledge of PanNENs' biology through novel findings, including higher-than-expected presence of germline mutations in 17% of sporadic tumours of no familial background, identification of novel mutational signatures and complex chromosomal rearrangements and a dysregulated epigenetic machinery. Integrated genomic studies have progressed the field by identifying the synergistic action between different molecular mechanisms, while holding the promise for deciphering disease heterogeneity. Although our understanding is far from being complete, these novel findings have provided the optimism of shaping the future of PanNEN research, ultimately leading to an era of precision medicine for NETs. Here, we recapitulate the existing knowledge on pancreatic neuroendocrine tumours (PanNETs) and discuss how recent, novel findings have furthered our understanding of these complex tumours.


Asunto(s)
Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética , Epigénesis Genética , Humanos , Tumores Neuroendocrinos/terapia , Neoplasias Pancreáticas/terapia
5.
Nat Med ; 25(3): 517-525, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30664780

RESUMEN

The molecular alterations that occur in cells before cancer is manifest are largely uncharted. Lung carcinoma in situ (CIS) lesions are the pre-invasive precursor to squamous cell carcinoma. Although microscopically identical, their future is in equipoise, with half progressing to invasive cancer and half regressing or remaining static. The cellular basis of this clinical observation is unknown. Here, we profile the genomic, transcriptomic, and epigenomic landscape of CIS in a unique patient cohort with longitudinally monitored pre-invasive disease. Predictive modeling identifies which lesions will progress with remarkable accuracy. We identify progression-specific methylation changes on a background of widespread heterogeneity, alongside a strong chromosomal instability signature. We observed mutations and copy number changes characteristic of cancer and chart their emergence, offering a window into early carcinogenesis. We anticipate that this new understanding of cancer precursor biology will improve early detection, reduce overtreatment, and foster preventative therapies targeting early clonal events in lung cancer.


Asunto(s)
Carcinoma in Situ/genética , Carcinoma de Células Escamosas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinogénesis/genética , Inestabilidad Cromosómica/genética , Estudios de Cohortes , Variaciones en el Número de Copia de ADN , Metilación de ADN/genética , Progresión de la Enfermedad , Epigenómica , Femenino , Perfilación de la Expresión Génica , Genómica , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Mutación
6.
JAMA Oncol ; 1(4): 476-85, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26181258

RESUMEN

IMPORTANCE: The utility of buccal cells as an epithelial source tissue for epigenome-wide association studies (EWASs) remains to be demonstrated. Given the direct exposure of buccal cells to potent carcinogens such as smoke, epigenetic changes in these cells may provide insights into the development of smoke-related cancers. OBJECTIVE: To perform an EWAS in buccal and blood cells to assess the relative effect of smoking on the DNA methylation (DNAme) patterns in these cell types and to test whether these DNAme changes are also seen in epithelial cancer. DESIGN, SETTING, AND PARTICIPANTS: In 2013, we measured DNAme at more than 480,000 CpG sites in buccal samples provided in 1999 by 790 women (all aged 53 years in 1999) from the United Kingdom Medical Research Council National Survey of Health and Development. This included matched blood samples from 152 women. We constructed a DNAme-based smoking index and tested its sensitivity and specificity to discriminate normal from cancer tissue in more than 5000 samples. MAIN OUTCOMES AND MEASURES: CpG sites whose DNAme level correlates with smoking pack-years, and construction of an associated sample-specific smoking index, which measures the mean deviation of DNAme at smoking-associated CpG sites from a normal reference. RESULTS: In a discovery set of 400 women, we identified 1501 smoking-associated CpG sites at a genome-wide significance level of P < 10-7, which were validated in an independent set of 390 women. This represented a 40-fold increase of differentially methylated sites in buccal cells compared with matched blood samples. Hypermethylated sites were enriched for bivalently marked genes and binding sites of transcription factors implicated in DNA repair and chromatin architecture (P < 10-10). A smoking index constructed from the DNAme changes in buccal cells was able to discriminate normal tissue from cancer tissue with a mean receiver operating characteristic area under the curve of 0.99 (range, 0.99-1.00) for lung cancers and of 0.91 (range, 0.71-1.00) for 13 other organs. The corresponding area under the curve of a smoking signature derived from blood cells was lower than that derived from buccal cells in 14 of 15 cancer types (Wilcoxon signed rank test, P = .001). CONCLUSIONS AND RELEVANCE: These data point toward buccal cells as being a more appropriate source of tissue than blood to conduct EWASs for smoking-related epithelial cancers.


Asunto(s)
Transformación Celular Neoplásica/genética , Metilación de ADN , Células Epiteliales/metabolismo , Pruebas Genéticas/métodos , Neoplasias Pulmonares/genética , Mucosa Bucal/metabolismo , Neoplasias Glandulares y Epiteliales/genética , Fumar/genética , Área Bajo la Curva , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Islas de CpG , Células Epiteliales/patología , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Persona de Mediana Edad , Mucosa Bucal/patología , Neoplasias Glandulares y Epiteliales/sangre , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Valor Predictivo de las Pruebas , Curva ROC , Factores de Riesgo , Fumar/efectos adversos , Reino Unido
8.
Science ; 345(6196): 1251343, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25082706

RESUMEN

Long interspersed nuclear element-1 (L1) retrotransposons are mobile repetitive elements that are abundant in the human genome. L1 elements propagate through RNA intermediates. In the germ line, neighboring, nonrepetitive sequences are occasionally mobilized by the L1 machinery, a process called 3' transduction. Because 3' transductions are potentially mutagenic, we explored the extent to which they occur somatically during tumorigenesis. Studying cancer genomes from 244 patients, we found that tumors from 53% of the patients had somatic retrotranspositions, of which 24% were 3' transductions. Fingerprinting of donor L1s revealed that a handful of source L1 elements in a tumor can spawn from tens to hundreds of 3' transductions, which can themselves seed further retrotranspositions. The activity of individual L1 elements fluctuated during tumor evolution and correlated with L1 promoter hypomethylation. The 3' transductions disseminated genes, exons, and regulatory elements to new locations, most often to heterochromatic regions of the genome.


Asunto(s)
Elementos Transponibles de ADN , Elementos de Nucleótido Esparcido Largo , Neoplasias/genética , Transducción Genética , Carcinogénesis/genética , Cromatina/química , Exones , Genoma Humano , Humanos , Mutagénesis Insercional , Translocación Genética
9.
Nat Commun ; 5: 3644, 2014 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-24714652

RESUMEN

Cancer evolves by mutation, with somatic reactivation of retrotransposons being one such mutational process. Germline retrotransposition can cause processed pseudogenes, but whether this occurs somatically has not been evaluated. Here we screen sequencing data from 660 cancer samples for somatically acquired pseudogenes. We find 42 events in 17 samples, especially non-small cell lung cancer (5/27) and colorectal cancer (2/11). Genomic features mirror those of germline LINE element retrotranspositions, with frequent target-site duplications (67%), consensus TTTTAA sites at insertion points, inverted rearrangements (21%), 5' truncation (74%) and polyA tails (88%). Transcriptional consequences include expression of pseudogenes from UTRs or introns of target genes. In addition, a somatic pseudogene that integrated into the promoter and first exon of the tumour suppressor gene, MGA, abrogated expression from that allele. Thus, formation of processed pseudogenes represents a new class of mutation occurring during cancer development, with potentially diverse functional consequences depending on genomic context.


Asunto(s)
Neoplasias/genética , Seudogenes/genética , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Seudogenes/fisiología
10.
Thorax ; 69(6): 548-57, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24550057

RESUMEN

BACKGROUND: Squamous cell carcinoma of the lung is a common cancer with 95% mortality at 5 years. These cancers arise from preinvasive lesions, which have a natural history of development progressing through increasing severity of dysplasia to carcinoma in situ (CIS), and in some cases, ending in transformation to invasive carcinoma. Synchronous preinvasive lesions identified at autopsy have been previously shown to be clonally related. METHODS: Using autofluorescence bronchoscopy that allows visual observation of preinvasive lesions within the upper airways, together with molecular profiling of biopsies using gene sequencing and loss-of-heterozygosity analysis from both preinvasive lesions and from intervening normal tissue, we have monitored individual lesions longitudinally and documented their visual, histological and molecular relationship. RESULTS: We demonstrate that rather than forming a contiguous field of abnormal tissue, clonal CIS lesions can develop at multiple anatomically discrete sites over time. Further, we demonstrate that patients with CIS in the trachea have invariably had previous lesions that have migrated proximally, and in one case, into the other lung over a period of 12 years. CONCLUSIONS: Molecular information from these unique biopsies provides for the first time evidence that field cancerisation of the upper airways can occur through cell migration rather than via local contiguous cellular expansion as previously thought. Our findings urge a clinical strategy of ablating high-grade premalignant airway lesions with subsequent attentive surveillance for recurrence in the bronchial tree.


Asunto(s)
Carcinoma in Situ , Carcinoma de Células Escamosas , Movimiento Celular , Neoplasias Pulmonares , Mutación , Lesiones Precancerosas , Neoplasias de la Tráquea , Adulto , Carcinoma in Situ/genética , Carcinoma in Situ/patología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Genes p53 , Humanos , Pérdida de Heterocigocidad , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Neoplasias de la Tráquea/genética , Neoplasias de la Tráquea/patología
11.
Elife ; 2: e00966, 2013 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-24151545

RESUMEN

Lineage tracing approaches have provided new insights into the cellular mechanisms that support tissue homeostasis in mice. However, the relevance of these discoveries to human epithelial homeostasis and its alterations in disease is unknown. By developing a novel quantitative approach for the analysis of somatic mitochondrial mutations that are accumulated over time, we demonstrate that the human upper airway epithelium is maintained by an equipotent basal progenitor cell population, in which the chance loss of cells due to lineage commitment is perfectly compensated by the duplication of neighbours, leading to "neutral drift" of the clone population. Further, we show that this process is accelerated in the airways of smokers, leading to intensified clonal consolidation and providing a background for tumorigenesis. This study provides a benchmark to show how somatic mutations provide quantitative information on homeostatic growth in human tissues, and a platform to explore factors leading to dysregulation and disease. DOI:http://dx.doi.org/10.7554/eLife.00966.001.


Asunto(s)
Células Madre/metabolismo , Procesos Estocásticos , Tráquea/metabolismo , Células Epiteliales/metabolismo , Humanos , Fumar/metabolismo , Fumar/patología , Tráquea/citología
12.
J Pathol ; 224(2): 153-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21506132

RESUMEN

The term 'field cancerization' is used to describe an epithelial surface that has a propensity to develop cancerous lesions, and in the case of the aerodigestive tract this is often as a result of chronic exposure to carcinogens in cigarette smoke 1, 2. The clinical endpoint is the development of multiple tumours, either simultaneously or sequentially in the same epithelial surface. The mechanisms underlying this process remain unclear; one possible explanation is that the epithelium is colonized by a clonal population of cells that are at increased risk of progression to cancer. We now address this possibility in a short case series, using individual genomic events as molecular biomarkers of clonality. In squamous lung cancer the most common genomic aberration is 3q amplification. We use a digital PCR technique to assess the clonal relationships between multiple biopsies in a longitudinal bronchoscopic study, using amplicon boundaries as markers of clonality. We demonstrate that clonality can readily be defined by these analyses and confirm that field cancerization occurs at a pre-invasive stage and that pre-invasive lesions and subsequent cancers are clonally related. We show that while the amplicon boundaries can be shared between different biopsies, the degree of 3q amplification and the internal structure of the 3q amplicon varies from lesion to lesion. Finally, in this small cohort, the degree of 3q amplification corresponds to clinical progression.


Asunto(s)
Bronquios/patología , Carcinoma de Células Escamosas/patología , Neoplasias Pulmonares/patología , Células Madre Neoplásicas/patología , Lesiones Precancerosas/patología , Adulto , Biopsia , Carcinoma de Células Escamosas/genética , Progresión de la Enfermedad , Femenino , Genómica/métodos , Humanos , Estudios Longitudinales , Neoplasias Pulmonares/genética , Microdisección/métodos , Lesiones Precancerosas/genética
13.
Biomarkers ; 13(7): 680-91, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19096962

RESUMEN

OBJECTIVES: Several clinical studies have indicated that the rates of invasive growth and metastatic disease in cancer depend on the degree of hypoxia, which is mediated by hypoxia-inducible factor 1 alpha (HIF-1alpha). To determine its potential role as a marker for prostate cancer (CaP) diagnosis, HIF-1alpha mRNA levels were measured in blood samples of patients diagnosed with different stages of prostatic disease. METHODS: HIF-1alpha mRNA levels were measured by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) and correlated with accurate clinicopathological data. Quantitative data were compared with serum prostate-specific antigen (PSA) measurements to determine variations in the accuracy of the CaP diagnosis. RESULTS: HIF-1alpha mRNA levels were significantly upregulated in patients with localized CaP (LocCaP; n=63; p<0.0001), compared with patients with no evidence of malignancy (NEOM) and benign prostatic hyperplasia (BPH) (n=35 for both patient groups combined). Receiver operator characteristic (ROC) curve analysis demonstrated that HIF-1alpha specificity for the NEOM/BPH diagnosis was 88.6%. Sensitivity for LocCaP was 74.6% with an overall diagnostic efficiency of 79.6%. Specificity of the NEOM diagnosis at PSA levels of 4.0 ng ml(-1) was 28.6% and sensitivity of the LocCap diagnosis was 65.1%, demonstrating a reduced overall diagnostic efficiency, compared with that given by HIF-1alpha measurements, of 52.0%. Levels of HIF-1alpha in patients with metastatic CaP (MetCaP; n=27)) were similar to those in the NEOM/BPH group. CONCLUSIONS: HIF-1alpha is upregulated early in CaP development with subsequent downregulation at later metastatic stages. This study demonstrates increased accuracy of early-stage disease diagnosis using HIF-1alpha qRT-PCR compared with serum PSA measurements. HIF-1alpha may therefore be a useful adjunct, together with other diagnostic markers used in relative qRT-PCR and current diagnostic techniques (including serum PSA and PSA velocity) to minimize unnecessary biopsies indicated by elevated serum PSA levels alone.


Asunto(s)
Detección Precoz del Cáncer , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias de la Próstata/diagnóstico , ARN Neoplásico/sangre , Biomarcadores de Tumor , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Metástasis de la Neoplasia , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , ARN Mensajero/sangre , Curva ROC , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Factores de Tiempo
14.
Biomarkers ; 12(5): 541-57, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17701752

RESUMEN

The use of serum prostate-specific antigen (PSA) measurements necessitates biopsies for accurate prostate cancer (CaP) diagnosis. Overall efficiency of accurate diagnosis, when PSA levels are used alone, is less than 60%. E2F3 was evaluated as an alternative biomarker using patient blood samples. Expression levels were measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and correlated with accurate clinicopathological data. Statistical analysis demonstrated significant differences in E2F3 expression levels (p<0.0001), and high levels of discrimination (receiver operator curve/area under curve analysis values (AUC) >0.88), in particular at early stages of disease development, between benign disease and localized CaP. Limited levels of discrimination were observed at the later stages of disease development, between localized and metastatic disease (p=0.076, AUC=0.633). A cut-off point of 0.34 with high specificity for benign disease (92.3%) and sensitivity for CaP diagnosis (81.0%) was identified. At this cut-off point, 85% patients were correctly diagnosed with either malignant or benign disease. This study demonstrates the strength of E2F3 as a potential marker for discriminating benign and malignant disease, addressing the current limitations of serum PSA measurements.


Asunto(s)
Biomarcadores de Tumor/sangre , Factor de Transcripción E2F3/genética , Neoplasias de la Próstata/sangre , ARN Mensajero/sangre , Proteína C-Reactiva/análisis , Línea Celular Tumoral , Cartilla de ADN/genética , ADN Complementario/química , ADN Complementario/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Antígeno Prostático Específico/sangre , Hiperplasia Prostática/sangre , Hiperplasia Prostática/diagnóstico , Hiperplasia Prostática/genética , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/genética , ARN Mensajero/genética , Curva ROC , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Temperatura de Transición
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...