Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Genet ; 9(4): 401-6, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7795646

RESUMEN

Overexpression of amplified genes is often associated with the acquisition of resistance to cancer therapeutic agents in vitro. We have identified a similar molecular mechanism in vivo for endocrine treatment failure in human prostate cancer which involves amplification of the androgen receptor (AR) gene. Comparative genomic hybridization shows that amplification of the Xq11-q13 region (the location), is common in tumours recurring during androgen deprivation therapy. We found high-level AR amplification in seven of 23 (30%) recurrent tumours, but in none of the specimens taken from the same patients prior to therapy. Our results suggest that AR amplification emerges during androgen deprivation therapy by facilitating tumour cell growth in low androgen concentrations.


Asunto(s)
Amplificación de Genes , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Anciano , Resistencia a Medicamentos/genética , Humanos , Hibridación Fluorescente in Situ/métodos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/terapia , Cromosoma X
2.
Nat Genet ; 20(2): 175-9, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9771711

RESUMEN

Over 200,000 new prostate cancer cases are diagnosed in the United States each year, accounting for more than 35% of all cancer cases affecting men, and resulting in 40,000 deaths annually. Attempts to characterize genes predisposing to prostate cancer have been hampered by a high phenocopy rate, the late age of onset of the disease and, in the absence of distinguishing clinical features, the inability to stratify patients into subgroups relative to suspected genetic locus heterogeneity. We previously performed a genome-wide search for hereditary prostate cancer (HPC) genes, finding evidence of a prostate cancer susceptibility locus on chromosome 1 (termed HPC1; ref. 2). Here we present evidence for the location of a second prostate cancer susceptibility gene, which by heterogeneity estimates accounts for approximately 16% of HPC cases. This HPC locus resides on the X chromosome (Xq27-28), a finding consistent with results of previous population-based studies suggesting an X-linked mode of HPC inheritance. Linkage to Xq27-28 was observed in a combined study population of 360 prostate cancer families collected at four independent sites in North America, Finland and Sweden. A maximum two-point lod score of 4.60 was observed at DXS1113, theta=0.26, in the combined data set. Parametric multipoint and non-parametric analyses provided results consistent with the two-point analysis. Significant evidence for genetic locus heterogeneity was observed, with similar estimates of the proportion of linked families in each separate family collection. Genetic mapping of the locus represents an important initial step in the identification of an X-linked gene implicated in the aetiology of HPC.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Neoplasias de la Próstata/genética , Cromosoma X , Adulto , Anciano , Anciano de 80 o más Años , Mapeo Cromosómico , Marcadores Genéticos , Genotipo , Humanos , Escala de Lod , Masculino , Persona de Mediana Edad , Linaje , Receptores Androgénicos/genética
3.
Nat Genet ; 30(2): 181-4, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11799394

RESUMEN

Although prostate cancer is the most common non-cutaneous malignancy diagnosed in men in the United States, little is known about inherited factors that influence its genetic predisposition. Here we report that germline mutations in the gene encoding 2'-5'-oligoadenylate(2-5A)-dependent RNase L (RNASEL) segregate in prostate cancer families that show linkage to the HPC1 (hereditary prostate cancer 1) region at 1q24-25 (ref. 9). We identified RNASEL by a positional cloning/candidate gene method, and show that a nonsense mutation and a mutation in an initiation codon of RNASEL segregate independently in two HPC1-linked families. Inactive RNASEL alleles are present at a low frequency in the general population. RNASEL regulates cell proliferation and apoptosis through the interferon-regulated 2-5A pathway and has been suggested to be a candidate tumor suppressor gene. We found that microdissected tumors with a germline mutation showed loss of heterozygosity and loss of RNase L protein, and that RNASEL activity was reduced in lymphoblasts from heterozyogous individuals compared with family members who were homozygous with respect to the wildtype allele. Thus, germline mutations in RNASEL may be of diagnostic value, and the 2-5A pathway might provide opportunities for developing therapies for those with prostate cancer.


Asunto(s)
Endorribonucleasas/genética , Mutación de Línea Germinal , Oncogenes , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Nucleótidos de Adenina/metabolismo , Secuencia de Bases , Estudios de Casos y Controles , Análisis Mutacional de ADN , ADN de Neoplasias/genética , Femenino , Ligamiento Genético , Heterocigoto , Homocigoto , Humanos , Pérdida de Heterocigocidad , Linfocitos/enzimología , Masculino , Oligorribonucleótidos/metabolismo , Linaje
4.
Nat Med ; 4(7): 844-7, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9662379

RESUMEN

Many genes and signalling pathways controlling cell proliferation, death and differentiation, as well as genomic integrity, are involved in cancer development. New techniques, such as serial analysis of gene expression and cDNA microarrays, have enabled measurement of the expression of thousands of genes in a single experiment, revealing many new, potentially important cancer genes. These genome screening tools can comprehensively survey one tumor at a time; however, analysis of hundreds of specimens from patients in different stages of disease is needed to establish the diagnostic, prognostic and therapeutic importance of each of the emerging cancer gene candidates. Here we have developed an array-based high-throughput technique that facilitates gene expression and copy number surveys of very large numbers of tumors. As many as 1000 cylindrical tissue biopsies from individual tumors can be distributed in a single tumor tissue microarray. Sections of the microarray provide targets for parallel in situ detection of DNA, RNA and protein targets in each specimen on the array, and consecutive sections allow the rapid analysis of hundreds of molecular markers in the same set of specimens. Our detection of six gene amplifications as well as p53 and estrogen receptor expression in breast cancer demonstrates the power of this technique for defining new subgroups of tumors.


Asunto(s)
Neoplasias de la Mama/genética , Técnicas Genéticas , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclina D1/genética , Ciclina D1/metabolismo , Femenino , Humanos , Técnicas para Inmunoenzimas , Hibridación Fluorescente in Situ , Ratones , Proteínas Oncogénicas v-myb , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Conejos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Proteínas Oncogénicas de Retroviridae/genética , Proteínas Oncogénicas de Retroviridae/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
5.
Science ; 258(5083): 818-21, 1992 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-1359641

RESUMEN

Comparative genomic hybridization produces a map of DNA sequence copy number as a function of chromosomal location throughout the entire genome. Differentially labeled test DNA and normal reference DNA are hybridized simultaneously to normal chromosome spreads. The hybridization is detected with two different fluorochromes. Regions of gain or loss of DNA sequences, such as deletions, duplications, or amplifications, are seen as changes in the ratio of the intensities of the two fluorochromes along the target chromosomes. Analysis of tumor cell lines and primary bladder tumors identified 16 different regions of amplification, many in loci not previously known to be amplified.


Asunto(s)
ADN de Neoplasias/genética , Neoplasias/genética , Hibridación de Ácido Nucleico , Mapeo Cromosómico , Sondas de ADN , Femenino , Fluoresceína-5-Isotiocianato , Colorantes Fluorescentes , Amplificación de Genes , Eliminación de Gen , Humanos , Hibridación Fluorescente in Situ , Masculino , Mutación , Oncogenes , Polimorfismo de Longitud del Fragmento de Restricción , Rodaminas , Células Tumorales Cultivadas
6.
Science ; 277(5328): 965-8, 1997 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-9252329

RESUMEN

Members of the recently recognized SRC-1 family of transcriptional coactivators interact with steroid hormone receptors to enhance ligand-dependent transcription. AIB1, a member of the SRC-1 family, was cloned during a search on the long arm of chromosome 20 for genes whose expression and copy number were elevated in human breast cancers. AIB1 amplification and overexpression were observed in four of five estrogen receptor-positive breast and ovarian cancer cell lines. Subsequent evaluation of 105 unselected specimens of primary breast cancer found AIB1 amplification in approximately 10 percent and high expression in 64 percent of the primary tumors analyzed. AIB1 protein interacted with estrogen receptors in a ligand-dependent fashion, and transfection of AIB1 resulted in enhancement of estrogen-dependent transcription. These observations identify AIB1 as a nuclear receptor coactivator whose altered expression may contribute to development of steroid-dependent cancers.


Asunto(s)
Neoplasias de la Mama/genética , Amplificación de Genes , Neoplasias Hormono-Dependientes/genética , Neoplasias Ováricas/genética , Receptores de Estrógenos/metabolismo , Secuencia de Aminoácidos , Mama/metabolismo , Neoplasias de la Mama/metabolismo , Cromosomas Humanos Par 20 , Clonación Molecular , Estradiol/metabolismo , Estradiol/farmacología , Femenino , Dosificación de Gen , Regulación Neoplásica de la Expresión Génica , Histona Acetiltransferasas , Humanos , Hibridación Fluorescente in Situ , Ligandos , Datos de Secuencia Molecular , Neoplasias Hormono-Dependientes/metabolismo , Coactivador 1 de Receptor Nuclear , Coactivador 2 del Receptor Nuclear , Neoplasias Ováricas/metabolismo , Receptores de Estrógenos/genética , Factores de Transcripción/genética , Activación Transcripcional , Transfección , Células Tumorales Cultivadas
7.
Science ; 274(5291): 1371-4, 1996 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-8910276

RESUMEN

Despite its high prevalence, very little is known regarding genetic predisposition to prostate cancer. A genome-wide scan performed in 66 high-risk prostate cancer families has provided evidence of linkage to the long arm of chromosome 1 (1q24-25). Analysis of an additional set of 25 North American and Swedish families with markers in this region resulted in significant evidence of linkage in the combined set of 91 families. The data provide strong evidence of a major prostate cancer susceptibility locus on chromosome 1.


Asunto(s)
Mapeo Cromosómico , Cromosomas Humanos Par 1 , Genes , Neoplasias de la Próstata/genética , Adulto , Anciano , Anciano de 80 o más Años , Repeticiones de Dinucleótido , Ligamiento Genético , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Humanos , Funciones de Verosimilitud , Masculino , Persona de Mediana Edad , América del Norte , Oncogenes , Linaje , Factores de Riesgo , Estadísticas no Paramétricas , Suecia
8.
Oncogene ; 26(26): 3846-56, 2007 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-17160014

RESUMEN

Serial analysis of gene expression followed by pathway analysis implicated the tight junction protein claudin-1 (CLDN1) in melanoma progression. Tight junction proteins regulate the paracellular transport of molecules, but staining of a tissue microarray revealed that claudin-1 was overexpressed in melanoma, and aberrantly expressed in the cytoplasm of malignant cells, suggesting a role other than transport. Indeed, melanoma cells in culture demonstrate no tight junction function. It has been shown that protein kinase C (PKC) can affect expression of claudin-1 in rat choroid plexus cells, and we observed a correlation between levels of activated PKC and claudin expression in our melanoma cells. To determine if PKC could affect the expression of CLDN1 in human melanoma, cells lacking endogenous claudin-1 were treated with 200 nM phorbol myristic acid (PMA). PKC activation by PMA caused an increase in CLDN1 transcription in 30 min, and an increase in claudin-1 protein by 12 h. Inhibition of PKC signaling in cells with high claudin-1 expression resulted in decreased claudin-1 expression. CLDN1 appears to contribute to melanoma cell invasion, as transient transfection of melanoma cells with CLDN1 increased metalloproteinase 2 (MMP-2) secretion and activation, and subsequently, motility of melanoma cells as demonstrated by wound-healing assays. Conversely, knockdown of CLDN1 by siRNA resulted in the inhibition of motility, as well as decreases in MMP-2 secretion and activation. These data implicate claudin-1 in melanoma progression.


Asunto(s)
Movimiento Celular/fisiología , Melanoma/patología , Proteínas de la Membrana/metabolismo , Invasividad Neoplásica/fisiopatología , Proteína Quinasa C/metabolismo , Western Blotting , Línea Celular Tumoral , Claudina-1 , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Metaloproteinasa 2 de la Matriz/metabolismo , Melanoma/metabolismo , Microscopía Confocal , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
9.
Br J Cancer ; 98(8): 1457-66, 2008 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-18349832

RESUMEN

Multiple genetic loci confer susceptibility to breast and ovarian cancers. We have previously developed a model (BOADICEA) under which susceptibility to breast cancer is explained by mutations in BRCA1 and BRCA2, as well as by the joint multiplicative effects of many genes (polygenic component). We have now updated BOADICEA using additional family data from two UK population-based studies of breast cancer and family data from BRCA1 and BRCA2 carriers identified by 22 population-based studies of breast or ovarian cancer. The combined data set includes 2785 families (301 BRCA1 positive and 236 BRCA2 positive). Incidences were smoothed using locally weighted regression techniques to avoid large variations between adjacent intervals. A birth cohort effect on the cancer risks was implemented, whereby each individual was assumed to develop cancer according to calendar period-specific incidences. The fitted model predicts that the average breast cancer risks in carriers increase in more recent birth cohorts. For example, the average cumulative breast cancer risk to age 70 years among BRCA1 carriers is 50% for women born in 1920-1929 and 58% among women born after 1950. The model was further extended to take into account the risks of male breast, prostate and pancreatic cancer, and to allow for the risk of multiple cancers. BOADICEA can be used to predict carrier probabilities and cancer risks to individuals with any family history, and has been implemented in a user-friendly Web-based program (http://www.srl.cam.ac.uk/genepi/boadicea/boadicea_home.html).


Asunto(s)
Neoplasias de la Mama/genética , Genes BRCA1 , Genes BRCA2 , Predisposición Genética a la Enfermedad , Mutación , Neoplasias Ováricas/genética , Adulto , Factores de Edad , Anciano , Neoplasias de la Mama/etiología , Femenino , Tamización de Portadores Genéticos , Humanos , Persona de Mediana Edad , Modelos Genéticos , Neoplasias Primarias Secundarias/etiología , Neoplasias Primarias Secundarias/genética , Neoplasias Ováricas/etiología
11.
Trends Genet ; 13(10): 405-9, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9351342

RESUMEN

Comparative genomic hybridization (CGH) provides a molecular cytogenetic approach for genome-wide scanning of differences in DNA sequence copy number. The technique is now attracting wide-spread interest, especially among cancer researchers. The rapidly expanding database of CGH publications already covers about 1500 tumors and is beginning to reveal genetic abnormalities that are characteristic of certain tumor types or stages of tumor progression. Six novel gene amplifications, as well as a locus for a cancer-predisposition syndrome, have been discovered based on CGH data. CGH has now been established as a first-line screening technique for cancer researchers and will serve as a basis for ongoing efforts to develop high-resolution next-generation genome scanning, such as the microarray technology.


Asunto(s)
Hibridación in Situ/métodos , Neoplasias/genética , Animales , Predicción , Amplificación de Genes , Humanos , Metástasis de la Neoplasia/genética , Neoplasias/diagnóstico , Neoplasias Experimentales/genética
12.
J Med Genet ; 42(7): 602-3, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15994883

RESUMEN

A recent report estimated the breast cancer risks in carriers of the three Ashkenazi founder mutations to be higher than previously published estimates derived from population based studies. In an attempt to confirm this, the breast and ovarian cancer risks associated with the three Ashkenazi founder mutations were estimated using families included in a previous meta-analysis of populatrion based studies. The estimated breast cancer risks for each of the founder BRCA1 and BRCA2 mutations were similar to the corresponding estimates based on all BRCA1 or BRCA2 mutations in the meta-analysis. These estimates appear to be consistent with the observed prevalence of the mutations in the Ashkenazi Jewish population.


Asunto(s)
Neoplasias de la Mama/genética , Genes BRCA1 , Genes BRCA2 , Heterocigoto , Mutación , Neoplasias Ováricas/genética , Adulto , Anciano , Neoplasias de la Mama/epidemiología , Femenino , Efecto Fundador , Humanos , Incidencia , Judíos/genética , Metaanálisis como Asunto , Persona de Mediana Edad , Neoplasias Ováricas/epidemiología , Penetrancia , Prevalencia , Medición de Riesgo
13.
J Natl Cancer Inst ; 84(14): 1109-14, 1992 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-1352359

RESUMEN

BACKGROUND: Recent evidence indicates that a subset of axillary node-negative (ANN) breast cancer patients can benefit from adjuvant therapy. Reliable prognostic markers are needed, however, to help clinicians identify these patients and arrive at more rational treatment decisions. PURPOSE: Mutations of the p53 tumor suppressor gene often result in overexpression of the p53 protein. In this study, we evaluated the prognostic significance of p53 protein overexpression in patients with ANN breast cancer. We also studied the association between the tumor cell proliferation rate and overexpression of the p53 and c-erbB-2 proteins, both of which have been implicated in cell cycle control. The c-erbB-2 protein is the product of the ERBB2 gene. METHODS: Two hundred eighty-nine ANN cases were randomly selected from a population-based cohort of patients who had not received any kind of adjuvant chemotherapy or endocrine therapy. Overexpression of the p53 and c-erbB-2 proteins was studied immunohistochemically in archival paraffin-embedded tumor samples, using the CM-1 polyclonal and the TAb 250 monoclonal antibodies, respectively. The tumor cell proliferation rate was measured as the S-phase fraction by DNA flow cytometry. Statistical analyses were performed using BMDP software. RESULTS: High-level p53 protein overexpression, found in 41 of the 289 tumors, was most common in tumors with high histologic grade, negative estrogen receptor status, c-erbB-2 protein overexpression, DNA index greater than 1.3, or high S-phase fraction. The lowest S-phase levels were found in tumors with neither p53 nor c-erbB-2 protein overexpression; the highest levels were seen in tumors showing overexpression of both proteins (P less than .0001). Both p53 and c-erbB-2 overexpression, as well as tumor size, had independent prognostic value in multivariate analysis. Eight-year survival of patients with p53 protein overexpression was 56% compared with 81% in patients with no overexpression (relative risk, 3.7; P less than .0001). If the S-phase fraction was included in a Cox regression analysis, however, only the tumor size and the S-phase fraction emerged as independent predictors of survival. CONCLUSIONS: Overexpression of the p53 and c-erbB-2 proteins indicates a high malignant potential in ANN breast cancer, but it is not a significant prognostic factor independent of the cell proliferation rate. The correlation between overexpression of these proteins and an increased S-phase fraction suggests that they may confer a proliferative advantage to cancer cells in vivo.


Asunto(s)
Neoplasias de la Mama/genética , Expresión Génica/fisiología , Genes p53/genética , Proteína p53 Supresora de Tumor/genética , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Ciclo Celular/fisiología , División Celular/fisiología , Femenino , Humanos , Inmunohistoquímica , Metástasis Linfática/diagnóstico , Adhesión en Parafina , Pronóstico , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/genética , Receptor ErbB-2 , Fase S/fisiología , Análisis de Supervivencia , Proteína p53 Supresora de Tumor/análisis
14.
J Natl Cancer Inst ; 84(11): 883-7, 1992 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-1350643

RESUMEN

BACKGROUND: Mutations in the p53 gene resulting in the accumulation of altered p53 proteins with prolonged half-life have been found in a large variety of human malignancies. PURPOSE: We studied the significance of p53 protein accumulation in prostatic carcinoma. METHODS: The material consisted of 137 paraffin-embedded, primary prostatic carcinomas. Accumulation of p53 protein was studied by immunohistochemical staining using a polyclonal p53-specific CM-1 antibody. Proliferation activity was determined by DNA flow cytometry and by immunohistochemical detection of proliferative cell nuclear antigen (PCNA) using a monoclonal PC10 antibody. RESULTS: Eight (6%) of the tumors showed intense p53 staining in more than 20% of the tumor cells, 15 (11%) had only lower level immunoreactivity, and 114 (83%) showed no staining. High-level p53 accumulation was associated with high histologic grade (P less than .001), DNA aneuploidy (P less than .05), and high cell proliferation rate as defined by flow cytometric S-phase analysis (P less than .01) or PCNA expression (P less than .01). High-level p53 accumulation predicted short, progression-free interval (P less than .01) and poor survival (P less than .001), with about a 12-fold relative risk of death as compared with p53-negative cases. Low-level p53 accumulation had no prognostic significance. CONCLUSIONS: Accumulation of p53 confers proliferative advantage for prostatic carcinoma cells and defines a small subgroup of highly malignant carcinomas.


Asunto(s)
Biomarcadores de Tumor/análisis , Genes p53 , Mutación , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteína p53 Supresora de Tumor/análisis , Anciano , Anticuerpos , ADN de Neoplasias/análisis , Citometría de Flujo , Estudios de Seguimiento , Humanos , Inmunohistoquímica , Masculino , Proteínas Nucleares/análisis , Pronóstico , Antígeno Nuclear de Célula en Proliferación , Estudios Retrospectivos , Factores de Tiempo , Proteína p53 Supresora de Tumor/genética
15.
J Natl Cancer Inst ; 92(15): 1252-9, 2000 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-10922410

RESUMEN

BACKGROUND: Studies by comparative genomic hybridization (CGH) have shown that chromosomal region 17q23 is amplified in up to 20% of primary breast cancers. We used microarray analyses to measure the expression levels of genes in this region and to explore their prognostic importance. METHODS: A microarray that contained 4209 complementary DNA (cDNA) clones was used to identify genes that are overexpressed in the MCF-7 breast cancer cell line as compared with normal mammary tissue. Fluorescence in situ hybridization was used to analyze the copy number of one overexpressed gene, ribosomal protein S6 kinase (S6K), and to localize it to the 17q23 region. Northern and western blot analyses were used to measure S6K gene and protein expression, and an enzymatic assay was used to measure S6K activity. Tumor tissue microarray analysis was used to study amplification of S6K and the HER-2 oncogene, another 17q-linked gene, and the relationship between amplification and prognosis was analyzed. The Kaplan-Meier method was used for data analysis, and the log-rank test was used for statistical analysis. All P values are two-sided. RESULTS: S6K was amplified and highly overexpressed in MCF-7 cells relative to normal mammary epithelium, and protein expression and enzyme activity were increased. S6K was amplified in 59 (8.8%) of 668 primary breast tumors, and a statistically significant association between amplification and poor prognosis (P =.0021) was observed. Amplification of both S6K and HER-2 implied particularly poor survival (P =.0001). CONCLUSIONS: The combination of CGH information with cDNA and tissue microarray analyses can be used to identify amplified and overexpressed genes and to evaluate the clinical implications of such genes and genomic rearrangements. S6K is likely to be one of the genes at 17q23 that is amplified during oncogenesis and may adversely affect the prognosis of patients with this amplification.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Cromosomas Humanos Par 17/genética , ADN de Neoplasias/análisis , Perfilación de la Expresión Génica/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Quinasas S6 Ribosómicas/metabolismo , Northern Blotting , Western Blotting , Mama/enzimología , ADN Complementario , Activación Enzimática , Femenino , Regulación Neoplásica de la Expresión Génica , Genes erbB-2/genética , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Pronóstico , Proteínas Quinasas S6 Ribosómicas/genética , Análisis de Supervivencia , Células Tumorales Cultivadas , Regulación hacia Arriba
16.
J Natl Cancer Inst ; 91(20): 1758-64, 1999 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-10528027

RESUMEN

BACKGROUND: The molecular mechanisms underlying the progression of prostate cancer during hormonal therapy have remained poorly understood. In this study, we developed a new strategy for the identification of differentially expressed genes in hormone-refractory human prostate cancer by use of a combination of complementary DNA (cDNA) and tissue microarray technologies. METHODS: Differences in gene expression between hormone-refractory CWR22R prostate cancer xenografts (human prostate cancer transplanted into nude mice) and a xenograft of the parental, hormone-sensitive CWR22 strain were analyzed by use of cDNA microarray technology. To validate the data from cDNA microarrays on clinical prostate cancer specimens, a tissue microarray of specimens from 26 prostates with benign prostatic hyperplasia, 208 primary prostate cancers, and 30 hormone-refractory local recurrences was constructed and used for immunohistochemical detection of protein expression. RESULTS: Among 5184 genes surveyed with cDNA microarray technology, expression of 37 (0.7%) was increased more than twofold in the hormone-refractory CWR22R xenografts compared with the CWR22 xenograft; expression of 135 (2.6%) genes was reduced by more than 50%. The genes encoding insulin-like growth factor-binding protein 2 (IGFBP2) and 27-kd heat-shock protein (HSP27) were among the most consistently overexpressed genes in the CWR22R tumors. Immunohistochemical analysis of tissue microarrays demonstrated high expression of IGFBP2 protein in 100% of the hormone-refractory clinical tumors, in 36% of the primary tumors, and in 0% of the benign prostatic specimens (two-sided P =.0001). Overexpression of HSP27 protein was demonstrated in 31% of the hormone-refractory tumors, in 5% of the primary tumors, and in 0% of the benign prostatic specimens (two-sided P =.0001). CONCLUSIONS: The combination of cDNA and tissue microarray technologies enables rapid identification of genes associated with progression of prostate cancer to the hormone-refractory state and may facilitate analysis of the role of the encoded gene products in the pathogenesis of human prostate cancer.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , ADN Complementario/efectos de los fármacos , ADN de Neoplasias/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Animales , ADN Complementario/análisis , ADN de Neoplasias/análisis , Proteínas de Choque Térmico/análisis , Proteínas de Choque Térmico/genética , Humanos , Inmunohistoquímica , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/análisis , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Masculino , Ratones , Ratones Desnudos , Recurrencia Local de Neoplasia , Hiperplasia Prostática/genética , Neoplasias de la Próstata/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo , Insuficiencia del Tratamiento
17.
Cancer Res ; 57(7): 1222-7, 1997 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-9102202

RESUMEN

BRCA1 and BRCA2 mutations confer increased risk for development of breast cancer, but a number of additional, currently largely unknown, somatic genetic defects must also accumulate in the breast epithelial cells before malignancy develops. To evaluate the nature of these additional somatic genetic defects, we performed a genome-wide survey by comparative genomic hybridization on breast cancers from 21 BRCA1 mutation carriers, 15 BRCA2 mutation carriers, and 55 unselected controls. The total number of genetic changes was almost two times higher in tumors from both BRCA1 and BRCA2 mutation carriers than in the control group. In BRCA1 tumors, losses of 5q (86%), 4q (81%), 4p (64%), 2q (40%), and 12q (40%) were significantly more common than in the control group (7-13%). BRCA2 tumors were characterized by a higher frequency of 13q (73%) and 6q (60%) losses and gains of 17q22-q24 (87%) and 20q13 (60%) as compared to the prevalence of these changes in the control group (12-18%). In conclusion, accumulation of somatic genetic changes during tumor progression may follow a unique pathway in individuals genetically predisposed to cancer, especially by the BRCA1 gene. Activation or loss of genes in the affected chromosomal regions may be selected for during tumor progression in cells lacking functional BRCA1 or BRCA2. Identification of such genes could provide targets for therapeutic intervention and early diagnosis.


Asunto(s)
Neoplasias de la Mama/genética , Genes BRCA1 , Mutación de Línea Germinal , Proteínas de Neoplasias/genética , Factores de Transcripción/genética , Adulto , Anciano , Anciano de 80 o más Años , Proteína BRCA2 , Carcinoma Ductal de Mama/genética , Deleción Cromosómica , Cromosomas Humanos Par 4 , Cromosomas Humanos Par 5 , Progresión de la Enfermedad , Femenino , Heterocigoto , Humanos , Persona de Mediana Edad , Hibridación de Ácido Nucleico , Polimorfismo Conformacional Retorcido-Simple
18.
Cancer Res ; 57(8): 1597-604, 1997 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-9108466

RESUMEN

To understand the genetic basis and clonal evolution underlying metastatic progression of human breast cancer in vivo, we analyzed the genetic composition of 29 primary breast carcinomas and their paired asynchronous metastases by comparative genomic hybridization and fluorescence in situ hybridization. The mean number of genetic changes by comparative genomic hybridization was 8.7 +/- 5.3 in primary tumors and 9.0 +/- 5.7 in their metastases. Although most of the genetic changes occurred equally often in the two groups, gains of the Xq12-q22 region were enriched in the metastases. According to a statistical analysis of shared genetic changes and breakpoints in paired specimens, 20 of the metastases (69%) showed a high degree of clonal relationship with the corresponding primary tumor, whereas the genetic composition of 9 metastases (31%) differed almost completely from that of the paired primary tumors. In both groups, however, chromosome X inactivation patterns suggested that the metastatic lesions originated from the same clone as the primary tumor. Fluorescence in situ hybridization analysis with probes specific to metastatic clones usually failed to find such cells in the primary tumor sample. In conclusion, detailed characterization of the in vivo progression pathways of metastatic breast cancer indicates that a linear progression model is unlikely to account for the progression of primary tumors to metastases. An early stem line clone apparently evolves independently in the primary tumor and its metastasis, eventually leading to multiple, genetically almost completely different, clones in the various tumor locations in a given patient. The resulting heterogeneity of metastatic breast cancer may underlie its poor responsiveness to therapy and explain why biomarkers of prognosis or therapy responsiveness measured exclusively from primary tumors give a restricted view of the biological properties of metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Aberraciones Cromosómicas , Adenocarcinoma/genética , Adenocarcinoma/patología , Adulto , Anciano , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/patología , Carcinoma Lobular/genética , Carcinoma Lobular/patología , Carcinoma Medular/genética , Carcinoma Medular/patología , Compensación de Dosificación (Genética) , Femenino , Humanos , Hibridación Fluorescente in Situ , Interfase , Persona de Mediana Edad , Metástasis de la Neoplasia
19.
Cancer Res ; 57(2): 314-9, 1997 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-9000575

RESUMEN

Progression of prostate cancer during endocrine therapy is a major clinical problem, the molecular mechanisms of which remain poorly understood. Amplification of the androgen receptor (AR) gene was recently described in recurrent prostate carcinomas from patients who had failed androgen deprivation therapy. To evaluate the hypothesis that amplification of the AR gene is a cause for the failure of androgen deprivation therapy in prostate cancer, we studied whether AR amplification leads to gene overexpression, whether the amplified AR gene is structurally intact, and whether tumors with AR amplification have distinct biological and clinical characteristics. Tumor specimens were collected from 54 prostate cancer patients at the time of a local recurrence following therapy failure. In 26 cases, paired primary tumor specimens from the same patients prior to therapy were also available. Fifteen (28%) of the recurrent therapy-resistant tumors, but none of the untreated primary tumors, contained AR gene amplification as determined by fluorescence in situ hybridization. According to single-stranded conformation polymorphism analysis, the AR gene was wild type in all but one of the 13 AR amplified cases studied. In one tumor, a presumed mutation in the hormone-binding domain at codon 674 leading to a Gly --> Ala substitution was found, but functional studies indicated that this mutation did not change the transactivational properties of the receptor. AR amplification was associated with a substantially increased level of mRNA expression of the gene by in situ hybridization. Clinicopathological correlations indicated that AR amplification was most likely to occur in tumors that had initially responded well to endocrine therapy and whose response duration was more than 12 months. Tumors that recurred earlier or those that showed no initial therapy response did not contain AR amplification. The median survival time after recurrence was two times longer for patients with AR amplification in comparison to those with no amplification (P = 0.03, Willcoxon-Breslow test). In conclusion, failure of conventional androgen deprivation therapy in prostate cancer may be caused by a clonal expansion of tumor cells that are able to continue androgen-dependent growth despite of the low concentrations of serum androgens. Amplification and the increased expression of a wild-type AR gene may play a key role in this process.


Asunto(s)
Amplificación de Genes/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/terapia , Receptores Androgénicos/genética , Anciano , Humanos , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Mutación Puntual , ARN Mensajero/metabolismo , Análisis de Supervivencia , Insuficiencia del Tratamiento
20.
Cancer Res ; 55(2): 342-7, 1995 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-7529134

RESUMEN

Genetic changes leading to the development of prostate cancer and factors that underlie the clinical progression of the disease are poorly characterized. Here, we used comparative genomic hybridization (CGH) to screen for DNA sequence copy number changes along all chromosomes in 31 primary and 9 recurrent uncultured prostate carcinomas. The aim of the study was to identify those chromosome regions that contain genes important for the development of prostate cancer and to identify genetic markers of tumor progression. CGH analysis indicated that 74% of primary prostate carcinoma showed DNA sequence copy number changes. Losses were 5 times more common than gains and most often involved 8p (32%), 13q (32%), 6q (22%), 16q (19%), 18q (19%), and 9p (16%). Allelic loss studies with 5 polymorphic microsatellite markers for 4 different chromosomes were done from 13 samples and showed a 76% concordance with CGH results. In local recurrences that developed during endocrine therapy, there were significantly more gains (P < 0.001) and losses (P < 0.05) of DNA sequences than in primary tumors, with gains of 8q (found in 89% of recurrences versus 6% of primary tumors), X (56% versus 0%), and 7 (56% versus 10%), as well as loss of 8p (78% versus 32%), being particularly often involved. In conclusion, our CGH results indicate that losses of several chromosomal regions are common genetic changes in primary tumors, suggesting that deletional inactivation of putative tumor suppressor genes in these chromosomal sites is likely to underlie development of prostate cancer. Furthermore, the pattern of genetic changes seen in recurrent tumors with the frequent gains of 7, 8q, and X suggests that the progression of prostate cancer and development of hormone-independent growth may have a distinct genetic basis. These chromosome aberrations may have diagnostic utility as markers of prostate cancer progression.


Asunto(s)
Deleción Cromosómica , Recurrencia Local de Neoplasia/genética , Neoplasias de la Próstata/genética , Genoma Humano , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino , Hibridación de Ácido Nucleico , Hiperplasia Prostática/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA