Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 194
Filtrar
Más filtros

Bases 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 ; 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
3.
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
4.
Br J Cancer ; 106(1): 99-106, 2012 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-22215106

RESUMEN

BACKGROUND: We have shown that a sodium ionophore monensin inhibits prostate cancer cell growth. A structurally related compound to monensin, salinomycin, was recently identified as a putative cancer stem cell inhibitor. METHODS: The growth inhibitory potential of salinomycin was studied in a panel of prostate cells. To get insights into the mechanism of action, a variety of assays such as gene expression and steroid profiling were performed in salinomycin-exposed prostate cancer cells. RESULTS: Salinomycin inhibited the growth of prostate cancer cells, but did not affect non-malignant prostate epithelial cells. Salinomycin impacted on prostate cancer stem cell functions as evidenced by reduced aldehyde dehydrogenase activity and the fraction of CD44(+) cells. Moreover, salinomycin reduced the expression of MYC, AR and ERG, induced oxidative stress as well as inhibited nuclear factor-κB activity and cell migration. Furthermore, profiling steroid metabolites revealed increased levels of oxidative stress-inducing steroids 7-ketocholesterol and aldosterone and decreased levels of antioxidative steroids progesterone and pregnenolone in salinomycin-exposed prostate cancer cells. CONCLUSION: Our results indicate that salinomycin inhibits prostate cancer cell growth and migration by reducing the expression of key prostate cancer oncogenes, inducing oxidative stress, decreasing the antioxidative capacity and cancer stem cell fraction.


Asunto(s)
División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Neoplasias de la Próstata/patología , Piranos/farmacología , Aldehído Deshidrogenasa/antagonistas & inhibidores , Western Blotting , Línea Celular Tumoral , Humanos , Masculino , FN-kappa B/metabolismo , Niclosamida/farmacología , Neoplasias de la Próstata/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores Androgénicos/efectos de los fármacos , Receptores Androgénicos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Terfenadina/farmacología
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
12.
Leukemia ; 32(3): 774-787, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28804127

RESUMEN

T-cell prolymphocytic leukemia (T-PLL) is a rare and aggressive neoplasm of mature T-cells with an urgent need for rationally designed therapies to address its notoriously chemo-refractory behavior. The median survival of T-PLL patients is <2 years and clinical trials are difficult to execute. Here we systematically explored the diversity of drug responses in T-PLL patient samples using an ex vivo drug sensitivity and resistance testing platform and correlated the findings with somatic mutations and gene expression profiles. Intriguingly, all T-PLL samples were sensitive to the cyclin-dependent kinase inhibitor SNS-032, which overcame stromal-cell-mediated protection and elicited robust p53-activation and apoptosis. Across all patients, the most effective classes of compounds were histone deacetylase, phosphoinositide-3 kinase/AKT/mammalian target of rapamycin, heat-shock protein 90 and BH3-family protein inhibitors as well as p53 activators, indicating previously unexplored, novel targeted approaches for treating T-PLL. Although Janus-activated kinase-signal transducer and activator of transcription factor (JAK-STAT) pathway mutations were common in T-PLL (71% of patients), JAK-STAT inhibitor responses were not directly linked to those or other T-PLL-specific lesions. Overall, we found that genetic markers do not readily translate into novel effective therapeutic vulnerabilities. In conclusion, novel classes of compounds with high efficacy in T-PLL were discovered with the comprehensive ex vivo drug screening platform warranting further studies of synergisms and clinical testing.


Asunto(s)
Antineoplásicos/farmacología , Biomarcadores de Tumor , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Ensayos Analíticos de Alto Rendimiento , Leucemia Prolinfocítica de Células T/genética , Mutación , Anciano , Anciano de 80 o más Años , Antineoplásicos/uso terapéutico , Ciclo Celular/genética , Línea Celular Tumoral , Aberraciones Cromosómicas , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Quinasas Janus/metabolismo , Leucemia Prolinfocítica de Células T/tratamiento farmacológico , Leucemia Prolinfocítica de Células T/metabolismo , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Oxazoles/farmacología , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Factores de Transcripción STAT/metabolismo , Tiazoles/farmacología
13.
Oncogene ; 25(52): 6997-7008, 2006 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-16715129

RESUMEN

Molecular mechanisms contributing to initiation and progression of head and neck squamous cell carcinoma are still poorly known. Numerous genetic alterations have been described, but molecular consequences of such alterations in most cases remain unclear. Here, we performed an integrated high-resolution microarray analysis of gene copy number and expression in 20 laryngeal cancer cell lines and primary tumors. Our aim was to identify genetic alterations that play a key role in disease pathogenesis and pinpoint genes whose expression is directly impacted by these events. Integration of DNA level data from array-based comparative genomic hybridization with RNA level information from oligonucleotide microarrays was achieved with custom-developed bioinformatic methods. High-level amplifications had a clear impact on gene expression. Across the genome, overexpression of 739 genes could be attributed to gene amplification events in cell lines, with 325 genes showing the same phenomenon in primary tumors including FADD and PPFIA1 at 11q13. The analysis of gene ontology and pathway distributions further pinpointed genes that may identify potential targets of therapeutic intervention. Our data highlight genes that may be critically important to laryngeal cancer progression and offer potential therapeutic targets.


Asunto(s)
Carcinoma de Células Escamosas/genética , Dosificación de Gen , Perfilación de la Expresión Génica/métodos , Neoplasias Laríngeas/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Línea Celular Tumoral , Expresión Génica , Humanos , Inmunohistoquímica
14.
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
15.
Eur J Pharm Sci ; 98: 30-39, 2017 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-27771514

RESUMEN

Urinary extracellular vesicles (EVs) are a promising source of biomarkers, which can be obtained in a non-invasive manner. However, the yield of EVs from urine samples may be insufficient for various analyses due to the entrapment of EVs by the Tamm-Horsfall protein (THP) meshwork. Here, we developed a simple dilution protocol to increase the urinary EV yield by disrupting the interaction between THP filaments and EVs with the help of alkaline pH and lowered ionic concentration. The integrity of the EVs and THP was assessed by electron microscopy. The effect of the protocol on the EV yield was quantified against an undiluted control by western blotting of four EV markers, nanoparticle tracking analysis and measuring of the RNA/miRNA concentration of the EV samples. The average EV yield from the dilution protocol was 2-7 fold the yield from the undiluted control i.e. increased by 130-624% as measured by western blotting and NTA. The yield increased most from samples with a high THP to EV ratio. The morphology and size range of the EVs were unaltered by the protocol. However, RNA/miRNA yields were the same as from the undiluted control and THP filaments could still be detected in EV samples. The dilution protocol, that we named KeepEX, provides a simple and efficient way to prevent loss of EVs thus increasing their yield from urine. Since KeepEX does not require individual adjustment of sample pH nor extra centrifugation steps, it could be used on its own or in combination with other EV purification protocols to improve EV isolation particularly from small urine volumes.


Asunto(s)
Vesículas Extracelulares , Orina/citología , Centrifugación , Vesículas Extracelulares/ultraestructura , Humanos , Concentración de Iones de Hidrógeno , Microscopía Electrónica de Transmisión , Proteínas/análisis , Trometamina/química
16.
Leukemia ; 31(1): 51-57, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27461063

RESUMEN

TCF3-PBX1 (E2A-PBX1) is a recurrent gene fusion in B-cell precursor acute lymphoblastic leukemia (BCP-ALL), which is caused by the translocation t(1;19)(q23;p13). TCF3-PBX1 BCP-ALL patients typically benefit from chemotherapy; however, many relapse and subsequently develop resistant disease with few effective treatment options. Mechanisms driving disease progression and therapy resistance have not been studied in TCF3-PBX1 BCP-ALL. Here, we aimed to identify novel treatment options for TCF3-PBX1 BCP-ALL by profiling leukemia cells from a relapsed patient, and determine molecular mechanisms underlying disease pathogenesis and progression. By drug-sensitivity testing of leukemic blasts from the index patient, control samples and TCF3-PBX1 positive and negative BCP-ALL cell lines, we identified the phosphatidylinositide 3-kinase delta (p110δ) inhibitor idelalisib as an effective treatment for TCF3-PBX1 BCP-ALL. This was further supported by evidence showing TCF3-PBX1 directly regulates expression of PIK3CD, the gene encoding p110δ. Other somatic mutations to TP53 and MTOR, as well as aberrant expression of CXCR4, may influence additional drug sensitivities specific to the index patient and accompanied progression of the disease. Our results suggest that idelalisib is a promising treatment option for patients with TCF3-PBX1 BCP-ALL, whereas other drugs could be useful depending on the genetic context of individual patients.


Asunto(s)
Proteínas de Fusión Oncogénica , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Purinas/farmacología , Quinazolinonas/farmacología , Adulto , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Proteínas de Fusión Oncogénica/fisiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Purinas/uso terapéutico , Quinazolinonas/uso terapéutico , Recurrencia
17.
Leukemia ; 31(5): 1187-1195, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27833094

RESUMEN

We sought to identify drugs that could counteract cytarabine resistance in acute myeloid leukemia (AML) by generating eight resistant variants from MOLM-13 and SHI-1 AML cell lines by long-term drug treatment. These cells were compared with 66 ex vivo chemorefractory samples from cytarabine-treated AML patients. The models and patient cells were subjected to genomic and transcriptomic profiling and high-throughput testing with 250 emerging and clinical oncology compounds. Genomic profiling uncovered deletion of the deoxycytidine kinase (DCK) gene in both MOLM-13- and SHI-1-derived cytarabine-resistant variants and in an AML patient sample. Cytarabine-resistant SHI-1 variants and a subset of chemorefractory AML patient samples showed increased sensitivity to glucocorticoids that are often used in treatment of lymphoid leukemia but not AML. Paired samples taken from AML patients before treatment and at relapse also showed acquisition of glucocorticoid sensitivity. Enhanced glucocorticoid sensitivity was only seen in AML patient samples that were negative for the FLT3 mutation (P=0.0006). Our study shows that development of cytarabine resistance is associated with increased sensitivity to glucocorticoids in a subset of AML, suggesting a new therapeutic strategy that should be explored in a clinical trial of chemorefractory AML patients carrying wild-type FLT3.


Asunto(s)
Citarabina/farmacología , Resistencia a Antineoplásicos , Glucocorticoides/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Adulto , Citarabina/uso terapéutico , Perfilación de la Expresión Génica , Humanos , Células Tumorales Cultivadas , Adulto Joven , Tirosina Quinasa 3 Similar a fms/genética
18.
Leukemia ; 31(5): 1048-1058, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27795554

RESUMEN

In our individualized systems medicine program, personalized treatment options are identified and administered to chemorefractory acute myeloid leukemia (AML) patients based on exome sequencing and ex vivo drug sensitivity and resistance testing data. Here, we analyzed how clonal heterogeneity affects the responses of 13 AML patients to chemotherapy or targeted treatments using ultra-deep (average 68 000 × coverage) amplicon resequencing. Using amplicon resequencing, we identified 16 variants from 4 patients (frequency 0.54-2%) that were not detected previously by exome sequencing. A correlation-based method was developed to detect mutation-specific responses in serial samples across multiple time points. Significant subclone-specific responses were observed for both chemotherapy and targeted therapy. We detected subclonal responses in patients where clinical European LeukemiaNet (ELN) criteria showed no response. Subclonal responses also helped to identify putative mechanisms underlying drug sensitivities, such as sensitivity to azacitidine in DNMT3A mutated cell clones and resistance to cytarabine in a subclone with loss of NF1 gene. In summary, ultra-deep amplicon resequencing method enables sensitive quantification of subclonal variants and their responses to therapies. This approach provides new opportunities for designing combinatorial therapies blocking multiple subclones as well as for real-time assessment of such treatments.


Asunto(s)
Células Clonales/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Antineoplásicos/farmacología , Secuencia de Bases , Monitoreo de Drogas , Variación Genética , Humanos , Leucemia Mieloide Aguda/genética , Terapia Molecular Dirigida , Medicina de Precisión
19.
Leukemia ; 31(2): 301-309, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27499136

RESUMEN

Inhibitors of B-cell lymphoma-2 (BCL-2) such as venetoclax (ABT-199) and navitoclax (ABT-263) are clinically explored in several cancer types, including acute myeloid leukemia (AML), to selectively induce apoptosis in cancer cells. To identify robust biomarkers for BCL-2 inhibitor sensitivity, we evaluated the ex vivo sensitivity of fresh leukemic cells from 73 diagnosed and relapsed/refractory AML patients, and then comprehensively assessed whether the responses correlated to specific mutations or gene expression signatures. Compared with samples from healthy donor controls (nonsensitive) and chronic lymphocytic leukemia (CLL) patients (highly sensitive), AML samples exhibited variable responses to BCL-2 inhibition. Strongest CLL-like responses were observed in 15% of the AML patient samples, whereas 32% were resistant, and the remaining exhibited intermediate responses to venetoclax. BCL-2 inhibitor sensitivity was associated with genetic aberrations in chromatin modifiers, WT1 and IDH1/IDH2. A striking selective overexpression of specific HOXA and HOXB gene transcripts were detected in highly BCL-2 inhibitor sensitive samples. Ex vivo responses to venetoclax showed significant inverse correlation to ß2-microglobulin expression and to a lesser degree to BCL-XL and BAX expression. As new therapy options for AML are urgently needed, the specific HOX gene expression pattern can potentially be used as a biomarker to identify venetoclax-sensitive AML patients for clinical trials.


Asunto(s)
Antineoplásicos/farmacología , Regulación Leucémica de la Expresión Génica , Genes Homeobox , Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Compuestos de Anilina/farmacología , Antineoplásicos/uso terapéutico , Biopsia , Médula Ósea/patología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Estudios de Casos y Controles , Línea Celular Tumoral , Análisis por Conglomerados , Resistencia a Antineoplásicos/genética , Exoma , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Isocitrato Deshidrogenasa/genética , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/tratamiento farmacológico , Familia de Multigenes , Mutación , Sulfonamidas/farmacología , Proteínas WT1/genética , Microglobulina beta-2/genética
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA