Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Neurooncol Adv ; 3(1): vdaa154, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33506200

RESUMEN

BACKGROUND: Mebendazole is an anthelmintic drug introduced for human use in 1971 that extends survival in preclinical models of glioblastoma and other brain cancers. METHODS: A single-center dose-escalation and safety study of mebendazole in 24 patients with newly diagnosed high-grade gliomas in combination with temozolomide was conducted. Patients received mebendazole in combination with adjuvant temozolomide after completing concurrent radiation plus temozolomide. Dose-escalation levels were 25, 50, 100, and 200 mg/kg/day of oral mebendazole. A total of 15 patients were enrolled at the highest dose studied of 200 mg/kg/day. Trough plasma levels of mebendazole were measured at 4, 8, and 16 weeks. RESULTS: Twenty-four patients (18 glioblastoma and 6 anaplastic glioma) were enrolled with a median age of 49.8 years. Four patients (at 200 mg/kg) developed elevated grade 3 alanine aminotransferase (ALT) and/or aspartate transaminase (AST) after 1 month, which reversed with lower dosing or discontinuation. Plasma levels of mebendazole were variable but generally increased with dose. Kaplan-Meier analysis showed a 21-month median overall survival with 41.7% of patients alive at 2 years and 25% at 3 and 4 years. Median progression-free survival (PFS) from the date of diagnosis for 17 patients taking more than 1 month of mebendazole was 13.1 months (95% confidence interval [CI]: 8.8-14.6 months) but for 7 patients who received less than 1 month of mebendazole PFS was 9.2 months (95% CI: 5.8-13.0 months). CONCLUSION: Mebendazole at doses up to 200 mg/kg demonstrated long-term safety and acceptable toxicity. Further studies are needed to determine mebendazole's efficacy in patients with malignant glioma.

2.
Oncotarget ; 8(46): 80124-80138, 2017 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-29113289

RESUMEN

PURPOSE: Glioblastoma is a deadly brain cancer with a median survival time of ∼15 months. Ionizing radiation plus the DNA alkylator temozolomide (TMZ) is the current standard therapy. PAC-1, a procaspase-3 activating small molecule, is blood-brain barrier penetrant and has previously demonstrated ability to synergize with diverse pro-apoptotic chemotherapeutics. We studied if PAC-1 could enhance the activity of TMZ, and whether addition of PAC-1 to standard treatment would be feasible in spontaneous canine malignant gliomas. EXPERIMENTAL DESIGN: Using cell lines and online gene expression data, we identified procaspase-3 as a potential molecular target for most glioblastomas. We investigated PAC-1 as a single agent and in combination with TMZ against glioma cells in culture and in orthotopic rodent models of glioma. Three dogs with spontaneous gliomas were treated with an analogous human glioblastoma treatment protocol, with concurrent PAC-1. RESULTS: Procaspase-3 is expressed in gliomas, with higher gene expression correlating with increased tumor grade and decreased prognosis. PAC-1 is cytotoxic to glioma cells in culture and active in orthotopic rodent glioma models. PAC-1 added to TMZ treatments in cell culture increases apoptotic death, and the combination significantly increases survival in orthotopic glioma models. Addition of PAC-1 to TMZ and radiation was well-tolerated in 3 out of 3 pet dogs with spontaneous glioma, and partial to complete tumor reductions were observed. CONCLUSIONS: Procaspase-3 is a clinically relevant target for treatment of glioblastoma. Synergistic activity of PAC-1/TMZ in rodent models and the demonstration of feasibility of the combined regime in canine patients suggest potential for PAC-1 in the treatment of glioblastoma.

3.
PLoS One ; 8(8): e70352, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936412

RESUMEN

Earlier, we reported that CTLA4 expression is inversely correlated with CD38 expression in chronic lymphocytic leukemia (CLL) cells. However, the specific role of CTLA4 in CLL pathogenesis remains unknown. Therefore, to elucidate the possible role of CTLA4 in CLL pathogenesis, CTLA4 was down-regulated in primary CLL cells. We then evaluated proliferation/survival in these cells using MTT, (3)H-thymidine uptake and Annexin-V apoptosis assays. We also measured expression levels of downstream molecules involved in B-cell proliferation/survival signaling including STAT1, NFATC2, c-Fos, c-Myc, and Bcl-2 using microarray, PCR, western blotting analyses, and a stromal cell culture system. CLL cells with CTLA4 down-regulation demonstrated a significant increase in proliferation and survival along with an increased expression of STAT1, STAT1 phosphorylation, NFATC2, c-Fos phosphorylation, c-Myc, Ki-67 and Bcl-2 molecules. In addition, compared to controls, the CTLA4-downregulated CLL cells showed a decreased frequency of apoptosis, which also correlated with increased expression of Bcl-2. Interestingly, CLL cells from lymph node and CLL cells co-cultured on stroma expressed lower levels of CTLA4 and higher levels of c-Fos, c-Myc, and Bcl-2 compared to CLL control cells. These results indicate that microenvironment-controlled-CTLA4 expression mediates proliferation/survival of CLL cells by regulating the expression/activation of STAT1, NFATC2, c-Fos, c-Myc, and/or Bcl-2.


Asunto(s)
Antígeno CTLA-4/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , ADP-Ribosil Ciclasa 1/metabolismo , Apoptosis , Linfocitos B/patología , Antígeno CTLA-4/deficiencia , Antígeno CTLA-4/genética , Proliferación Celular , Supervivencia Celular , Regulación hacia Abajo/genética , Silenciador del Gen , Humanos , Leucemia Linfocítica Crónica de Células B/diagnóstico , Pronóstico , ARN Interferente Pequeño/genética , Microambiente Tumoral
4.
PLoS One ; 7(10): e44372, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23056179

RESUMEN

Glioblastoma multiforme (GBM) is the most common intracranial cancer but despite recent advances in therapy the overall survival remains about 20 months. Whole genome exon sequencing studies implicate mutations in the receptor tyrosine kinase pathways (RTK) for driving tumor growth in over 80% of GBMs. In spite of various RTKs being mutated or altered in the majority of GBMs, clinical studies have not been able to demonstrate efficacy of molecular targeted therapies using tyrosine kinase inhibitors in GBMs. Activation of multiple downstream signaling pathways has been implicated as a possible means by which inhibition of a single RTK has been ineffective in GBM. In this study, we sought a combination of approved drugs that would inhibit in vitro and in vivo growth of GBM oncospheres. A combination consisting of gefitinib and sunitinib acted synergistically in inhibiting growth of GBM oncospheres in vitro. Sunitinib was the only RTK inhibitor that could induce apoptosis in GBM cells. However, the in vivo efficacy testing of the gefitinib and sunitinib combination in an EGFR amplified/PTEN wild type GBM xenograft model revealed that gefitinib alone could significantly improve survival in animals whereas sunitinib did not show any survival benefit. Subsequent testing of the same drug combination in a different syngeneic glioma model that lacked EGFR amplification but was more susceptible to sunitinib in vitro demonstrated no survival benefit when treated with gefitinib or sunitinib or the gefitinib and sunitinib combination. Although a modest survival benefit was obtained in one of two animal models with EGFR amplification due to gefitinib alone, the addition of sunitinib, to test our best in vitro combination therapy, did not translate to any additional in vivo benefit. Improved targeted therapies, with drug properties favorable to intracranial tumors, are likely required to form effective drug combinations for GBM.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Femenino , Gefitinib , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Immunoblotting , Indoles/administración & dosificación , Indoles/farmacología , Concentración 50 Inhibidora , Ratones , Ratones Desnudos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Pirroles/administración & dosificación , Pirroles/farmacología , Quinazolinas/administración & dosificación , Quinazolinas/farmacología , Ratas Endogámicas F344 , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Sunitinib , Resultado del Tratamiento
5.
Mol Cancer ; 10: 17, 2011 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-21314958

RESUMEN

BACKGROUND: Glioblastoma Multiforme (GBM) is the most common and invasive astrocytic tumor associated with dismal prognosis. Treatment for GBM patients has advanced, but the median survival remains a meager 15 months. In a recent study, 20,000 genes from 21 GBM patients were sequenced that identified frequent mutations in ion channel genes. The goal of this study was to determine whether ion channel mutations have a role in disease progression and whether molecular targeting of ion channels is a promising therapeutic strategy for GBM patients. Therefore, we compared GBM patient survival on the basis of presence or absence of mutations in calcium, potassium and sodium ion transport genes. Cardiac glycosides, known sodium channel inhibitors, were then tested for their ability to inhibit GBM cell proliferation. RESULTS: Nearly 90% of patients showed at least one mutation in ion transport genes. GBM patients with mutations in sodium channels showed a significantly shorter survival compared to patients with no sodium channel mutations, whereas a similar comparison based on mutational status of calcium or potassium ion channel mutations showed no survival differences. Experimentally, targeting GBM cells with cardiac glycosides such as digoxin and ouabain demonstrated preferential cytotoxicity against U-87 and D54 GBM cells compared to non-tumor astrocytes (NTAs). CONCLUSIONS: These pilot studies of GBM patients with sodium channel mutations indicate an association with a more aggressive disease and significantly shorter survival. Moreover, inhibition of GBM cells by ion channel inhibitors such as cardiac glycosides suggest a therapeutic strategy with relatively safe drugs for targeting GBM ion channel mutations. Key Words: glioblastoma multiforme, ion channels, mutations, small molecule inhibitors, cardiac glycosides.


Asunto(s)
Neoplasias Encefálicas/genética , Glioblastoma/genética , Mutación/genética , Canales de Sodio/genética , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/patología , Canales de Calcio/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Digoxina/farmacología , Glioblastoma/patología , Humanos , Masculino , Persona de Mediana Edad , Ouabaína/farmacología , Canales de Potasio/genética , Análisis de Supervivencia
6.
Cancer Res ; 70(22): 8981-7, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21045145

RESUMEN

Mutation at the R132 residue of isocitrate dehydrogenase 1 (IDH1), frequently found in gliomas and acute myelogenous leukemia, creates a neoenzyme that produces 2-hydroxyglutarate (2-HG) from α-ketoglutarate (α-KG). We sought to therapeutically exploit this neoreaction in mutant IDH1 cells that require α-KG derived from glutamine. Glutamine is converted to glutamate by glutaminase and further metabolized to α-KG. Therefore, we inhibited glutaminase with siRNA or the small molecule inhibitor bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide (BPTES) and found slowed growth of glioblastoma cells expressing mutant IDH1 compared with those expressing wild-type IDH1. Growth suppression of mutant IDH1 cells by BPTES was rescued by adding exogenous α-KG. BPTES inhibited glutaminase activity, lowered glutamate and α-KG levels, and increased glycolytic intermediates while leaving total 2-HG levels unaffected. The ability to selectively slow growth in cells with IDH1 mutations by inhibiting glutaminase suggests a unique reprogramming of intermediary metabolism and a potential therapeutic strategy.


Asunto(s)
Glutaminasa/metabolismo , Isocitrato Deshidrogenasa/metabolismo , Mutación , Western Blotting , Línea Celular Transformada , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Glioma/genética , Glioma/metabolismo , Glioma/patología , Ácido Glutámico/metabolismo , Glutaminasa/antagonistas & inhibidores , Glutaminasa/genética , Glutaratos/metabolismo , Humanos , Isocitrato Deshidrogenasa/genética , Ácidos Cetoglutáricos/metabolismo , Ácidos Cetoglutáricos/farmacología , Interferencia de ARN , Sulfuros/farmacología , Tiadiazoles/farmacología , Factores de Tiempo
7.
Int J Oncol ; 37(3): 645-53, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20664933

RESUMEN

Umbilical cord blood cells (UCBC) are a rich source of immature immune effector and accessory cells, including dendritic cells. UCBC-derived cytotoxic T lymphocytes (CTLs) generated against human breast cancer or neuroblastoma have shown an increased tumor-specific cytotoxicity compared to peripheral blood (PB)-derived CTLs. The precise mechanism of this increased cytotoxicity is not known. Since dendritic cells (DCs) play a central role in the immunostimulation, we compared the ultrastructure and antigen presenting nature of DCs from UCBC, PB and bone marrow (BM) at various stages of maturation using scanning and transmission electron microscopy as well as fluorescent microscopy to elucidate the mechanism underlying the increased cytotoxicity of UCBC-derived CTLs. DCs were examined for their immunophenotype nuclear morphology, dendritic processes and cytoplasmic endosomal vesicles after 0, 3, 7 and 10 days in culture with antigen priming on day 6. Results showed that there were smaller and more vesicles in UCB-DCs compared to DCs from the other two sources, while the endosomal vesicles in PB-DCs were heterogenous in size. The antigen processing ability of the UCB-DCs showed an increase in antigen-positive endosomes compared to PB-DCs as determined by the fluorescent microscopy. Thus, our results provided the comparative analyses of DCs from cord blood, peripheral blood and bone marrow, and suggested that UCBC-DCs might have better antigen presenting ability leading to increased CTL-mediated antitumor cytotoxicity.


Asunto(s)
Células de la Médula Ósea/inmunología , Sangre Fetal/inmunología , Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Presentación de Antígeno/inmunología , Células de la Médula Ósea/ultraestructura , Línea Celular Tumoral , Citotoxicidad Inmunológica/inmunología , Células Dendríticas/inmunología , Sangre Fetal/citología , Humanos , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Linfocitos T Citotóxicos/ultraestructura
8.
J Neurooncol ; 96(2): 169-79, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19609742

RESUMEN

Glioblastoma Multiforme (GBM) is a malignant brain cancer that develops after accumulating genomic DNA damage that often includes gene amplifications and/or deletions. These copy number changes can be a critical step in brain tumor development. To evaluate glioblastoma genomic copy number changes, we determined the genome-wide copy number alterations in 31 GBMs. Illumina Bead Arrays were used to assay 22 GBMs and Digital Karyotyping was used on 8 GBM cell lines and one primary sample. The common amplifications we observed for all 31 samples was GLI/CDK4 (22.6%), MDM2 (12.9%) and PIK3C2B/MDM4 (12.9%). In the 22 GBM tumors, EGFR was amplified in 22.7% of surgical biopsies. The most common homozygously deleted region contained CDKN2A/CDKN2B (p15 and p16) occurring in 29% of cases. This data was compiled and compared to published array CGH studies of 456 cases of GBMs. Pooling our Illumina data with published studies yielded these average amplification rates: EGFR-35.7%, GLI/CDK4-13.4%, MDM2-9.2%, PIK3C2B/MDM4-7.7%, and PDGFRA-7.7%. The CDKN2A/CDKN2B locus was deleted in 46.4% of the combined cases. This study provides a larger assessment of amplifications and deletions in glioblastoma patient populations and shows that several different copy number technologies can produce similar results. The main pathways known to be involved in GBM tumor formation such as p53 control, growth signaling, and cell cycle control are all represented by amplifications or deletions of critical pathway genes. This information is potentially important for formulating targeted therapy in glioblastoma and for planning genomic studies.


Asunto(s)
Neoplasias Encefálicas/genética , Amplificación de Genes/fisiología , Glioblastoma/genética , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Cromosomas Humanos/genética , Femenino , Dosificación de Gen , Perfilación de la Expresión Génica/métodos , Genoma Humano , Humanos , Cariotipificación/métodos , Masculino , Persona de Mediana Edad , Modelos Moleculares , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Eliminación de Secuencia/genética
9.
J Neurooncol ; 94(3): 359-66, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19340397

RESUMEN

The Fibroblast Growth Factor (FGF) signaling pathway is reported to stimulate glioblastoma (GBM) growth. In this work we evaluated the effect of FGF2, FGF receptor (FGFR), and small molecule inhibition on GBM cells grown in traditional media, or cultured directly in stem-cell media. These lines each expressed the FGFR1, FGFR3 and FGFR4 receptors. Addition of FGF2 ligand showed significant growth stimulation in 8 of 10 cell lines. Disruption of FGF signaling by a neutralizing FGF2 monoclonal antibody and FGFR1 suppression by RNA interference both partially inhibited cell proliferation. Growth inhibition was temporally correlated with a reduction in MAPK signaling. A receptor tyrosine kinase inhibitor with known FGFR/VEGFR activity, PD173074, showed reproducible growth inhibition. Possible mechanisms of growth suppression by PD173074 were implicated by reduced phosphorylation of AKT and MAPK, known oncogenic signal transducers. Subsequent reduction in the cyclin D1, cyclin D2 and CDK4 cell cycle regulators was also observed. Our results indicate that FGF signaling pathway inhibition as a monotherapy will slow, but not arrest growth of glioblastoma cells.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Glioblastoma/metabolismo , Glioblastoma/fisiopatología , Transducción de Señal/fisiología , Anticuerpos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factores de Crecimiento de Fibroblastos/inmunología , Factores de Crecimiento de Fibroblastos/farmacología , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Pirimidinas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Radioinmunoensayo/métodos , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Transfección/métodos
10.
Mol Cancer Res ; 6(12): 1928-36, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19074837

RESUMEN

B-cell chronic lymphocytic leukemia (B-CLL) is characterized by an accumulation of neoplastic B cells due to their resistance to apoptosis and increased survival. Among various factors, the tumor microenvironment is known to play a role in the regulation of cell proliferation and survival of many cancers. However, it remains unclear how the tumor microenvironment contributes to the increased survival of B-CLL cells. Therefore, we studied the influence of bone marrow stromal cell-induced hedgehog (Hh) signaling on the survival of B-CLL cells. Our results show that a Hh signaling inhibitor, cyclopamine, inhibits bone marrow stromal cell-induced survival of B-CLL cells, suggesting a role for Hh signaling in the survival of B-CLL cells. Furthermore, gene expression profiling of primary B-CLL cells (n = 48) indicates that the expression of Hh signaling molecules, such as GLI1, GLI2, SUFU, and BCL2, is significantly increased and correlates with disease progression of B-CLL patients with clinical outcome. In addition, SUFU and GLI1 transcripts, as determined by real-time PCR, are significantly overexpressed and correlate with adverse indicators of clinical outcome in B-CLL patients, such as cytogenetics or CD38 expression. Furthermore, selective down-regulation of GLI1 by antisense oligodeoxynucleotides (GLI1-ASO) results in decreased BCL2 expression and cell survival, suggesting that GLI1 may regulate BCL2 and, thereby, modulate cell survival in B-CLL. In addition, there was significantly increased apoptosis of B-CLL cells when cultured in the presence of GLI1-ASO and fludarabine. Together, these results reveal that Hh signaling is important in the pathogenesis of B-CLL and, hence, may be a potential therapeutic target.


Asunto(s)
Proteínas Hedgehog/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Leucemia Linfocítica Crónica de Células B/fisiopatología , Células del Estroma/citología , Células del Estroma/fisiología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Comunicación Celular/efectos de los fármacos , Comunicación Celular/fisiología , División Celular/efectos de los fármacos , División Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Oligodesoxirribonucleótidos Antisentido/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Transcripción/genética , Vidarabina/análogos & derivados , Vidarabina/farmacología , Proteína con Dedos de Zinc GLI1
11.
Int Immunopharmacol ; 8(9): 1257-63, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18602072

RESUMEN

Previous studies from this laboratory have characterized RAW117-P murine large cell B-cell lymphoma and its in vivo selected highly malignant and liver metastatic RAW117-H10 subline for their biological and biochemical properties. In this study, to understand the molecular basis of low and high metastatic behavior of these variant sublines, we have investigated the molecular phenotypes of these cells using differential display techniques and cDNA array analysis. Differential display analysis indicated a significant difference in expression of several genes between these two metastatic variant lymphoma cells. Further analyses of these cells using microarray showed an increased expression of several genes including uPAR1, CRE-BP1, Chop-10, IGF, insulin-like growth factor-IA, STAT6, Cyclin-D1, Cyclin-E, ERBB-3, Alpha NGF, Kruppel-like factor LKLF, (P)19INK4 in metastatic RAW117-H10 cells compared to parental RAW117-P cells. On the other hand, MIP1beta, CD14 antigen, Cathepsin B and MOD are expressed more in RAW117-P cells compared to RAW117-H10 cells. Differential expression of the selected genes was confirmed using semiquantitative RT-PCR techniques. The combination of plasminogen activator and its receptor and IGF-like growth factors, cell cycle regulatory molecules and transcription factors might provide an ideal environment for RAW117-H10 cells to metastasize to distant organs and colonize. Thus these results identify certain differentially expressed genes that are involved in the metastatic properties of these lymphoma cells and lay foundation for further in depth analyses to use this information to develop therapy for metastatic lymphoma.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Linfoma de Células B/genética , Animales , Línea Celular Tumoral , Células Cultivadas , Cartilla de ADN , ADN Complementario/biosíntesis , ADN Complementario/genética , Linfoma de Células B/patología , Ratones , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Mol Cancer Ther ; 7(6): 1450-60, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18524848

RESUMEN

Mantle cell lymphoma (MCL) has one of the worst clinical outcomes among the B-cell lymphomas, with a median survival of only 3 to 4 years. Therefore, a better understanding of the underlying mechanisms that regulate MCL proliferation/survival is needed to develop an effective therapy. Because sonic hedgehog (Shh)-GLI signaling has been shown to be important in the proliferation and survival of several cancers, and no such information is available for MCL, this study was undertaken. Our results show that the molecules associated with Shh-GLI signaling, such as PTCH and SMO receptors, and GLI1 and GLI2 target transcription factors were expressed in the human MCL cell lines and primary MCL cells from patients. Perturbation of this signaling in the presence of exogenous Shh/cyclopamine significantly (P < 0.001) influenced the proliferation of JVM2 MCL cells. Furthermore, down-regulation of GLI transcription factors using antisense oligonucleotides not only resulted in significantly (P < 0.001) decreased proliferation of the MCL cells but also significantly (P < 0.05) increased their susceptibility to chemotherapeutic drug, doxorubicin. Also, down-regulation of GLI decreased cyclin D1 and BCL2 transcript levels, which suggests that these key molecules might be regulated by GLI in MCL. Thus, our results indicate a significant role for Shh-GLI signaling in the proliferation of MCL, and molecular targeting of GLI is a potential therapeutic approach to improve the treatment for MCL.


Asunto(s)
Proteínas Hedgehog/metabolismo , Linfoma de Células del Manto/terapia , Transducción de Señal , Factores de Transcripción/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Doxorrubicina/farmacología , Regulación Neoplásica de la Expresión Génica/genética , Proteínas Hedgehog/genética , Humanos , Linfoma de Células del Manto/genética , Linfoma de Células del Manto/patología , Modelos Biológicos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética , Proteína con Dedos de Zinc GLI1
13.
J Neuroimmune Pharmacol ; 2(2): 202-12, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18040845

RESUMEN

Tumors of the nervous system, including neuroblastoma and glioblastoma, are difficult to treat with current therapies. Despite the advances in cancer therapeutics, the outcomes in these patients remain poor and, therefore, new modalities are required. Recent literature demonstrates that cytotoxic effector cells can effectively kill tumors of the nervous system. In addition, we have previously shown that umbilical cord blood (UCB) contains precursors of antitumor cytotoxic effector cells. Therefore, to evaluate the antitumor potential of UCB-derived effector cells, studies were designed to compare the in vitro and in vivo antitumor effects of UCB- and peripheral blood (PB)-derived antigen-nonspecific and antigen-specific effector cells against tumors of the nervous system. Mononuclear cells (MNCs) from UCB were used to generate both interleukin-2 (IL-2)-activated killer (LAK) cells and tumor-specific cytotoxic T lymphocytes (CTLs). UCB-derived LAK cells showed a significant in vitro cytotoxicity against IMR-32, SK-NMC, and U-87 human neuroblastoma and glioblastoma, respectively. In addition, the CTLs generated using dendritic cells primed with IMR-32 tumor cell lysate showed a selective cytotoxicity in vitro against IMR-32 cells, but not against U-87 or MDA-231 cells. Furthermore, treatment of SCID mice bearing IMR-32 neuroblastoma with tumor-specific CTLs resulted in a significant (p < 0.01) inhibition of tumor growth and increased overall survival. Thus, these results demonstrate the potential of UCB-derived effector cells against human neuroblastoma and warrant further preclinical studies.


Asunto(s)
Sangre Fetal/citología , Sangre Fetal/inmunología , Inmunoterapia/métodos , Neuroblastoma/inmunología , Neuroblastoma/terapia , Animales , Línea Celular Tumoral , Células Cultivadas , Citotoxicidad Inmunológica/inmunología , Sangre Fetal/trasplante , Glioblastoma/inmunología , Glioblastoma/terapia , Humanos , Células Asesinas Activadas por Linfocinas/inmunología , Células Asesinas Activadas por Linfocinas/trasplante , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/trasplante , Ratones , Ratones SCID , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/trasplante
14.
Clin Cancer Res ; 13(18 Pt 1): 5295-304, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17875758

RESUMEN

PURPOSE: In B-cell chronic lymphocytic leukemia (CLL), high CD38 expression has been associated with unfavorable clinical course, advanced disease, resistance to therapy, shorter time to first treatment, and shorter survival. However, the genes associated with CLL patient subgroups with high and low CD38 expression and their potential role in disease progression is not known. EXPERIMENTAL DESIGN: To identify the genes associated with the clinical disparity in CLL patients with high versus low CD38 expression, transcriptional profiles were obtained from CLL cells from 39 different patients using oligonucleotide microarray. Gene expression was also compared between CLL cells and B cells from healthy individuals. RESULTS: Gene expression analysis identified 76 differentially expressed genes in CD38 high versus low groups. Out of these genes, HEM1, CTLA4, and MNDA were selected for further studies and their differential expression was confirmed by real-time PCR. HEM1 overexpression was associated with poor outcome, whereas the overexpression of CTLA4 and MNDA was associated with good outcome. Down-regulation of HEM1 expression in patient CLL cells resulted in a significant increase in their susceptibility to fludarabine-mediated killing. In addition, when gene expression patterns in CD38 high and low CLL cells were compared with normal B-cell profiles, ATM expression was found to be significantly lower in CD38 high compared with CD38 low CLL as confirmed by real-time reverse transcription-PCR. CONCLUSIONS: These results identify the possible genes that may be involved in cell proliferation and survival and, thus, determining the clinical behavior of CLL patients expressing high or low CD38.


Asunto(s)
ADP-Ribosil Ciclasa 1/genética , Regulación Leucémica de la Expresión Génica , Genes Relacionados con las Neoplasias , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/mortalidad , Antígenos CD/genética , Antígenos de Diferenciación/genética , Antígenos de Diferenciación Mielomonocítica/genética , Proteínas de la Ataxia Telangiectasia Mutada , Antígeno CTLA-4 , Proteínas de Ciclo Celular/genética , Proliferación Celular , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Humanos , Leucemia Linfocítica Crónica de Células B/patología , Proteínas de la Membrana/genética , Pronóstico , Proteínas Serina-Treonina Quinasas/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética
15.
Int J Mol Med ; 20(4): 461-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17786276

RESUMEN

In B-cell chronic lymphocytic leukemia (CLL), Rai stage, immunoglobulin gene mutational status, chromosomal abnormalities, CD38 and ZAP-70 expression were used as prognostic markers. In this study, to understand the molecular basis of chromosomal abnormalities leading to tumor progression, 90 CLL patients were grouped into poor prognosis (with 11q deletion and trisomy 12) and good prognosis (with normal karyotype and 13q deletion) and their clinical outcome was assessed. Gene expression profiles of 35 CLL samples with poor outcome (11q deletion, n=9; trisomy 12, n=5) and good outcome (13q deletion, n=13; normal karyotype, n=8) were analyzed using oligonucleotide microarray. Significance analysis of microarray (SAM) identified 27 differentially expressed genes between these two subgroups with significant overexpression of ATF5 and underexpression of CDC16, PCDH8, SLAM, MNDA and ATF2 in CLL patients with poor outcome. ATF5 gene expression in CLL was further studied because of its role in the regulation of cell cycle progression/differentiation and apoptosis. The overexpression of ATF5 was confirmed by real-time PCR using 39 CLL samples from the poor and good outcome groups. ATF5 was significantly (p<0.001) overexpressed in the poor outcome group. Furthermore, ATF5 expression was significantly higher in the 11q deletion as well as trisomy 12 group alone compared to the 13q deletion and normal karyotype groups. ATF5 overexpression was also associated with significantly (p=0.04) shorter time to treatment. Similarly, expression of five underexpressed genes also correlated with longer time to treatment. Thus, this report demonstrates that ATF5 may be one of the key genes involved in increased proliferation and survival in 11q deletion or trisomy 12, whereas CD16, CD86, SLAM, MNDA and ATF2 may be involved in the decreased proliferation of CLL cells with 13q deletion or normal karyotype.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 12/genética , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Trisomía/genética , Factores de Transcripción Activadores/metabolismo , Análisis por Conglomerados , Análisis Citogenético , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias , Humanos , Leucemia Linfocítica Crónica de Células B/diagnóstico , Leucemia Linfocítica Crónica de Células B/terapia , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Tiempo , Resultado del Tratamiento
16.
Cancer Genet Cytogenet ; 172(2): 120-6, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17213020

RESUMEN

B-cell chronic lymphocytic leukemia (B-CLL) is the most common B-cell leukemia among older populations in Western countries. The clinical course of B-CLL is heterogeneous: in some patients the disease course is indolent, in others it is aggressive. The B-CLL subgroups with chromosome 11q23 deletion have been associated with aggressive disease course involving ATM deletion, extensive bulky lymphadenopathy (BLA), and inferior clinical outcome. Using real-time reverse transcriptase-polymerase chain reaction, we found that ATM was consistently underexpressed in B-CLL patients with BLA, irrespective of 11q23 deletion status. In addition, B-CLL patients who presented with BLA had a significantly shorter time to treatment (2 months) than did patients without BLA (74 months). Moreover, gene expression analysis in B-CLL patients with and without BLA revealed differences in expression for genes involved in apoptosis, cell cycle, and cell adhesion. These results indicate an association between BLA and reduced expression of ATM, suggesting a role for ATM in disease progression in B-CLL.


Asunto(s)
Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Deleción Cromosómica , Cromosomas Humanos Par 11/genética , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Regulación hacia Abajo/genética , Regulación Leucémica de la Expresión Génica , Leucemia Linfocítica Crónica de Células B/genética , Enfermedades Linfáticas/genética , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética , Proteínas de la Ataxia Telangiectasia Mutada , Adhesión Celular/genética , Ciclo Celular/genética , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Femenino , Humanos , Leucemia Linfocítica Crónica de Células B/diagnóstico , Leucemia Linfocítica Crónica de Células B/patología , Enfermedades Linfáticas/diagnóstico , Enfermedades Linfáticas/patología , Masculino , Persona de Mediana Edad , Pronóstico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Supresoras de Tumor/antagonistas & inhibidores
17.
Breast Cancer Res Treat ; 83(1): 15-23, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14997051

RESUMEN

The Her2/neu oncogene encodes a transmembrane protein with homology to the epidermal growth factor receptor. Overexpression of this gene contributes to the aggressiveness of breast cancer and poor prognosis. Therefore, Her2/neu is an ideal target molecule for generating effective cytotoxic T lymphocytes (CTLs) against breast cancers. This study reports on the generation of Her2/neu-specific CTL from umbilical cord blood mononuclear cells (UCBC) using dendritic cells primed with Her2/neu-derived peptide (KIFGSLAFL, E75) for immunostimulation. The CTLs showed specific cytotoxicity to Her2/neu high expressing MDA-453 but not toward Her2/neu low expressing MDA-231 human breast cancer cells. Similarly generated CTLs stimulated with irrelevant peptide pulsed dendritic cells did not show significant cytotoxicity towards breast cancer targets. The phenotypes of cells in culture showed high percentage of CD3+, CD4+ and CD8+T cells as determined by flow cytometry. However, the antibody mediated blocking assay demonstrated that only HLA-Class I restricted CD8+ cells are involved in the cytotoxicity. Furthermore, in vivo studies showed that treatment of SCID mice bearing MDA-453 tumor with Her2/neu-specific CTLs resulted in significant inhibition of tumor growth compared to untreated tumor bearing control mice. These results demonstrate that human umbilical cord blood mononuclear cells are a good source for generating Her2/neu-specific CTLs against human breast cancer both in vitro and in vivo.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias de la Mama/inmunología , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/farmacología , Linfocitos T Citotóxicos/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Células Dendríticas , Femenino , Sangre Fetal , Citometría de Flujo , Humanos , Leucocitos Mononucleares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA