Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Ann Oncol ; 31(8): 1040-1045, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32387456

RESUMEN

BACKGROUND: Cell entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) depends on binding of the viral spike (S) proteins to angiotensin-converting enzyme 2 and on S protein priming by TMPRSS2. Inhibition of TMPRSS2 may work to block or decrease the severity of SARS-CoV-2 infections. Intriguingly, TMPRSS2 is an androgen-regulated gene that is up-regulated in prostate cancer where it supports tumor progression and is involved in a frequent genetic translocation with the ERG gene. First- or second-generation androgen-deprivation therapies (ADTs) decrease the levels of TMPRSS2. Here we put forward the hypothesis that ADTs may protect patients affected by prostate cancer from SARS-CoV-2 infections. MATERIALS AND METHODS: We extracted data regarding 9280 patients (4532 males) with laboratory-confirmed SARS-CoV-2 infection from 68 hospitals in Veneto, one of the Italian regions that was most affected by the coronavirus disease 2019 (COVID-19) pandemic. The parameters used for each COVID-19-positive patient were sex, hospitalization, admission to intensive care unit, death, tumor diagnosis, prostate cancer diagnosis, and ADT. RESULTS: There were evaluable 9280 SARS-CoV-2-positive patients in Veneto on 1 April 2020. Overall, males developed more severe complications, were more frequently hospitalized, and had a worse clinical outcome than females. Considering only the Veneto male population (2.4 million men), 0.2% and 0.3% of non-cancer and cancer patients, respectively, tested positive for SARS-CoV-2. Comparing the total number of SARS-CoV-2-positive cases, prostate cancer patients receiving ADT had a significantly lower risk of SARS-CoV-2 infection compared with patients who did not receive ADT (OR 4.05; 95% CI 1.55-10.59). A greater difference was found comparing prostate cancer patients receiving ADT with patients with any other type of cancer (OR 4.86; 95% CI 1.88-12.56). CONCLUSION: Our data suggest that cancer patients have an increased risk of SARS-CoV-2 infections compared with non-cancer patients. However, prostate cancer patients receiving ADT appear to be partially protected from SARS-CoV-2 infections.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Betacoronavirus , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/prevención & control , Vigilancia de la Población , Neoplasias de la Próstata/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , COVID-19 , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/epidemiología , Humanos , Italia/epidemiología , Masculino , Persona de Mediana Edad , Neumonía Viral/diagnóstico , Neumonía Viral/epidemiología , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/epidemiología , Factores de Riesgo , SARS-CoV-2
2.
Invest New Drugs ; 35(6): 766-772, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28317088

RESUMEN

Purpose To establish a recommended phase II dose (RP2D) for the oral smoothened inhibitor sonidegib in combination with paclitaxel; secondary objectives include evaluation of safety, tolerability, markers of Hedgehog (Hh) signaling and preliminary antitumor activity. Methods Patients with advanced solid tumors were enrolled in cohorts of escalating sonidegib dose levels (400mg, 600mg and 800mg orally, once daily on days 1-28) in combination with paclitaxel 80 mg/m2 on days 1, 8 and 15 in 4-weekly cycles. Dose-limiting toxicities (DLTs) were assessed using CTCAE v4. Once the RP2D was defined, patients with advanced ovarian carcinoma were treated at this dose level in an expansion phase. Biomarkers of Hh signaling were assessed by immunohistochemistry in archival tissue and antitumor activity evaluated using RECIST 1.1. Results 18 patients were treated: 3 at 400 mg, 3 at 600 mg and 12 at 800 mg sonidegib. Only one patient treated at 800 mg presented a DLT (prolonged neutropenia resulting in failure to receive 75% of the planned sonidegib dose). However, 4 of 12 patients treated at 800 mg had their sonidegib dose reduced for toxicity after cycle 1. Hh biomarker (SHH, Patched, SMO and GLI1) staining did not correlate with clinical activity. Best response was partial response in 3 patients (2 ovarian, 1 breast cancer) and stable disease >4 cycles in 3 patients (2 ovarian, 1 anal cancer). Conclusions The combination of sonidegib and paclitaxel is tolerable and evidence of antitumor activity was identified. The RP2D of sonidegib was 800 mg in combination with paclitaxel 80mg/m2.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Receptor Smoothened/antagonistas & inhibidores , Administración Oral , Anciano , Biomarcadores de Tumor , Compuestos de Bifenilo/administración & dosificación , Relación Dosis-Respuesta a Droga , Femenino , Estudios de Seguimiento , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/metabolismo , Neoplasias/patología , Paclitaxel/administración & dosificación , Pronóstico , Piridinas/administración & dosificación
3.
Cancer Gene Ther ; 20(5): 326-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23618947

RESUMEN

Therapeutic gene silencing is a promising approach for treatment of cancer. Despite substantial efforts, however, only few such therapeutic methods have been clinically tested. The heterogeneity in gene expression profiles among malignant tissues and the dynamic control of gene expression in individual tumors makes identifying universal and effective targets a challenge. Further development of gene silencing therapy requires new approaches to comprehend and manage gene expression in cancer cells. In this study, we proposed and evaluated experimentally a new approach to design multi-gene silencing therapy. Using a simplified model of gene expression control, we show that genes commonly regulated by the same microRNA represent optimal combinations of targets for small hairpin RNA/small interfering RNA-based gene silencing. The proposed method of target gene selection and co-silencing can be explored as an algorithm for personalized cancer gene therapy.


Asunto(s)
Expresión Génica/genética , Silenciador del Gen , Terapia Genética/métodos , MicroARNs/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/terapia , Femenino , Perfilación de la Expresión Génica , Humanos , MicroARNs/metabolismo , Neoplasias Ováricas/metabolismo , Interferencia de ARN
4.
Oncogene ; 32(18): 2315-24, 2324.e1-4, 2013 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-22733135

RESUMEN

By integrating gene profiling and immunohistochemical data with functional experiments in cell lines in this study we show for the first time that doublecortin (DCX) domain containing 2 (DCDC2), a protein belonging to the DCX family and involved in neuronal cell migration, is aberrantly expressed in prostate tumors whereas absent in normal prostate. Furthermore, in patients treated with radical prostatectomy, high levels of DCDC2 RNA were significantly associated with increased biochemical relapse (LogRank Mantel-Cox=0.012). Mechanistically, we found that the ETS transcription factor ESE3/EHF, which is expressed in normal prostate and frequently lost in prostate tumors, maintained DCDC2 repressed by binding to a novel identified ETS binding site in the gene promoter. Consistently, in prostate tumors and in cellular models of gain and loss of ESE3/EHF, the expression of DCDC2 and ESE3/EHF were inversely correlated. In prostate cancer cells, DCDC2 colocalized with microtubules and promoted cell migration and resistance to the microtubule-targeting drug taxol. Collectively, this study establishes DCDC2 as a novel ESE3/EHF oncogenic target in prostate cancer. These findings may be relevant for the clinical management of prostate cancer as DCDC2 may signal tumors more prone to relapse and resistant to taxol treatment.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteínas Asociadas a Microtúbulos/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Sitios de Unión , Movimiento Celular/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Paclitaxel/farmacología , Regiones Promotoras Genéticas , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/cirugía , Valores de Referencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Moduladores de Tubulina/farmacología
5.
Oncogene ; 31(46): 4878-87, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22330138

RESUMEN

Epigenetic silencing of tumour suppressor genes is an important mechanism involved in cell transformation and tumour progression. The Set and RING-finger-associated domain-containing protein UHRF1 might be an important link between different epigenetic pathways. Here, we report that UHRF1 is frequently overexpressed in human prostate tumours and has an important role in prostate cancer pathogenesis and progression. Analysis of human prostate cancer samples by microarrays and immunohistochemistry showed increased expression of UHRF1 in about half of the cases. Moreover, UHRF1 expression was associated with reduced overall survival after prostatectomy in patients with organ-confined prostate tumours (P < 0.0001). UHRF1 expression was negatively correlated with several tumour suppressor genes and positively with the histone methyltransferase (HMT) EZH2 both in prostate tumours and cell lines. UHRF1 knockdown reduced proliferation, clonogenic capability and anchorage-independent growth of prostate cancer cells. Depletion of UHRF1 resulted in reactivation of several tumour suppressor genes. Gene reactivation upon UHRF1 depletion was associated with changes in histone H3K9 methylation, acetylation and DNA methylation, and impaired binding of the H3K9 HMT Suv39H1 to the promoter of silenced genes. Co-immunoprecipitation experiments showed direct interaction between UHRF1 and Suv39H1. Our data support the notion that UHRF1, along with Suv39H1 and DNA methyltransferases, contributes to epigenetic gene silencing in prostate tumours. This could represent a parallel and convergent pathway to the H3K27 methylation catalyzed by EZH2 to synergistically promote inactivation of tumour suppressor genes. Deregulated expression of UHRF1 is involved in the prostate cancer pathogenesis and might represent a useful marker to distinguish indolent cancer from those at high risk of lethal progression.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Acetilación , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Metilación de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Genes Supresores de Tumor , Células HEK293 , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Inmunoprecipitación/métodos , Masculino , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas , Neoplasias de la Próstata/patología , Ubiquitina-Proteína Ligasas
6.
Curr Cancer Drug Targets ; 11(8): 954-65, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21861840

RESUMEN

Castration-resistant prostate cancer remains incurable and a major cause of mortality worldwide. The absence of effective therapeutic approaches for advanced prostate cancer has led to an intensive search for novel treatments. Emerging nanomedical approaches have shown promising results, in vitro and in vivo, in improving drug distribution and bioavailability, tumor penetration and in limiting toxicity. Nanoscaled carriers bearing finely controlled size and surface properties such as liposomes, dendrimers and nanoparticles have been developed for successful passive and active tumortargeting. Enhanced pharmacokinetics of nanotherapeutics, through improved target delivery and prolonged tissue halflife provides optimal drug delivery that is tumor-specific. Tumor-targeting may be improved through ligand directed delivery systems binding to tumor-specific surface receptors improving cellular uptake through receptor-mediated endocytosis. Recently published data have provided pre-clinical evidence showing the potential of active-targeted nanotherapeutics in prostate cancer therapy; unfortunately, only a few of these therapies have translated into early phase clinical trials development. Hence, progress of active-targeted nanotherapy improving efficiency of site-specific drug delivery is a critical challenge in future clinical treatment of prostate cancer. Exploring specific prostate cell-surface antigens or receptor overexpression may elaborate promising strategies for future therapeutic design. This review presents an overview of some new strategies for prostate cancer active-targeting nanotherapeutics.


Asunto(s)
Antineoplásicos/administración & dosificación , Portadores de Fármacos/administración & dosificación , Terapia Molecular Dirigida , Nanoestructuras , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Portadores de Fármacos/uso terapéutico , Humanos , Masculino , Nanomedicina/métodos , Nanomedicina/tendencias , Nanoestructuras/química
7.
Eur J Cancer ; 44(13): 1829-34, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18640031

RESUMEN

BACKGROUND: The combination of a proteasome inhibitor with a taxane has potential clinical synergism that prompted a clinical test. PATIENTS AND METHODS: The maximum tolerated dose (MTD) and recommended dose (RD) of intravenous (i.v.) Bortezomib (B) (days 1, 4, 8, 11) and i.v. Paclitaxel (PTX) (days 1, 8) every 3 weeks was evaluated in patients with advanced solid tumours. The RD was tested in patients with breast, ovarian and prostate cancer. At the RD, microarray analysis of transcriptional profiles was carried out before and after the first dosing in peripheral blood mononuclear cells (PBMC). RESULTS: Thirty-one patients were enrolled and 22 were treated at the RD that corresponded to B 1.3mg/m(2) and PTX 100mg/m(2). The main toxicity was cumulative peripheral neuropathy (76% of patients; grade 3-4 in 9%) that required treatment discontinuation in six patients, followed by diarrhoea (55%) and fatigue (41%). Nine partial responses (30%) were observed (three breast cancer, four ovary, two prostate patients). Significant (p<0.05) and consistent changes (>70% of patients) in transcriptome were observed. CONCLUSIONS: The incidence of peripheral neuropathy and the anti-tumour activity comparable to that of single-agent PTX do not support further development of this regimen.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias de la Próstata/tratamiento farmacológico , Adulto , Anciano , Ácidos Borónicos/administración & dosificación , Ácidos Borónicos/efectos adversos , Bortezomib , Femenino , Humanos , Infusiones Intravenosas , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Paclitaxel/administración & dosificación , Paclitaxel/efectos adversos , Pirazinas/administración & dosificación , Pirazinas/efectos adversos , Resultado del Tratamiento
8.
Oncogene ; 27(20): 2877-85, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18037958

RESUMEN

Deregulated expression of ETS transcription factors has emerged as an important event in prostate cancer pathogenesis. Here we show that the expression of epithelial-specific ETS (ESE)-3 factor is frequently reduced at the RNA and protein level in prostate cancer clinical samples compared to normal prostate. In PC3 and DU145 cells, ESE-3 was silenced by methylation of an evolutionarily conserved CpG site in its promoter and treatment with 5-aza-2'-deoxycytidine restored its expression. In a prostate epithelial cell transformation model, methylation of this site was inversely correlated with ESE-3 expression and occurred only in Ras-transformed and tumorigenic cells and not in normal and immortalized cells suggesting that ESE-3 silencing was functionally linked to oncogenic transformation. Consistent with a tumor suppressor function, re-expression of ESE-3 in prostate cancer cells inhibited clonogenic survival and induced apoptotic cell death. ESE-3 increased the level of procaspase-3, a key element in the apoptotic cascade. This effect was mediated at the transcriptional level by direct binding of ESE-3 to the caspase-3 promoter. Collectively, our findings implicate ESE-3 as a candidate tumor suppressor in prostate cancer. Decreased expression of ESE-3 may result in loss of important regulatory mechanisms in prostate epithelial cells and contribute to the pathogenesis of prostate cancer.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/biosíntesis , Adulto , Anciano , Línea Celular Tumoral , Silenciador del Gen/fisiología , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata/genética , Factores de Transcripción/biosíntesis , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología
9.
Nucleic Acids Res ; 30(12): 2701-9, 2002 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12060688

RESUMEN

Altered expression of c-myc is implicated in pathogenesis and progression of many human cancers. Triple helix-forming oligonucleotides (TFOs) directed to a polypurine/polypyrimidine sequence in a critical regulatory region near the c-myc P2 promoter have been shown to inhibit c-myc transcription in vitro and in cells. However, these guanine-rich TFOs had moderate binding affinity and required high concentrations for activity. The 23 bp myc P2 sequence is split equally into AT- and GC-rich tracts. Gel mobility analysis of a series of short TFOs directed in parallel and anti-parallel orientation to the purine strand of each tract showed that only parallel CT and anti-parallel GT TFOs formed stable triplex on the AT- and GC-rich tracts, respectively. A novel full-length GTC TFO was designed to bind simultaneously in parallel and anti-parallel orientation to the polypurine strand. Gel-shift and footprinting assays showed that the new TFO formed a triple helix in physiological conditions with significantly higher affinity than an anti-parallel TFO. Protein-binding assays showed that 1 microM GTC TFO inhibited binding of nuclear transcription factors to the P2 promoter sequence. The novel TFO can be developed into a potent antigene agent, and its design strategy applied to similar genomic sequences, thus expanding the TFO repertoire.


Asunto(s)
ADN/síntesis química , ADN/farmacología , Genes myc , Oligodesoxirribonucleótidos/farmacología , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/genética , Secuencia Rica en At , Secuencia de Bases , Citosina/química , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Exodesoxirribonucleasas/química , Secuencia Rica en GC , Guanina/química , Humanos , Oligodesoxirribonucleótidos/síntesis química , Oligodesoxirribonucleótidos/metabolismo , Timidina/química , Células Tumorales Cultivadas
10.
Cancer Chemother Pharmacol ; 48(3): 188-96, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11592339

RESUMEN

PURPOSE: Topotecan is a topoisomerase I inhibitor with demonstrated anticancer activity in preclinical and clinical studies. The purpose of the present study was to evaluate drug-drug interactions in therapeutic regimens that would combine topotecan with microtubule-interfering agents, such as Taxol and vinblastine. METHODS: The cytotoxic activities of various drug combinations and schedules of administration were measured in a colon cancer cell line using the MTT assay. Western blot and flow cytometry were performed to determine the effects of Taxol and vinblastine on topoisomerase I and Bcl-xL protein levels and cell cycle distribution. RESULTS: Brief incubation of colon cancer cells with low concentrations of either Taxol or vinblastine increased the efficacy of a subsequent treatment with topotecan. Preincubation of cells with vinblastine or Taxol reduced by 10- to 40-fold the concentration of topotecan necessary to induce a 50% decrease in cell survival. The effects were maximal when the cells were treated for 5 h with microtubule-interfering agents and then incubated for 19 h in drug-free medium before the addition of topotecan. Under these conditions, both Taxol and vinblastine caused an increase in topoisomerase I protein levels, fraction of S phase cells, and extent of Bcl-xL phosphorylation immediately prior to the addition of topotecan. All these factors may contribute to the increased efficacy of topotecan observed with sequential therapy. CONCLUSION: Combinations of topotecan and microtubule-interfering agents result in synergistic anticancer activity when the drugs are administered sequentially. The promising preclinical data presented here encourage clinical testing of these drug combinations using a sequential schedule of administration.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Microtúbulos/efectos de los fármacos , Paclitaxel/farmacología , Topotecan/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Vinblastina/farmacología , Western Blotting , Ciclo Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , ADN-Topoisomerasas de Tipo I/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Formazáns , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sales de Tetrazolio , Inhibidores de Topoisomerasa I , Células Tumorales Cultivadas/metabolismo , Proteína bcl-X
11.
Biochem Pharmacol ; 62(1): 101-10, 2001 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-11377401

RESUMEN

Previous studies have demonstrated that cytosine arabinoside (araC) induces an accumulation of Okazaki fragments, while fludarabine (FaraA) inhibits Okazaki fragment synthesis. We extended these observations in the present study to provide insights into various mechanisms by which these anticancer drugs affect DNA replication and induce genomic instability in human CEM leukemia cells. Neither araC nor FaraA induced a detectable amount of re-replicated DNA in S-phase cells, which indicated that drug-induced alterations in Okazaki fragment synthesis were not accompanied by DNA re-replication. Synthesis on both leading and lagging DNA strands within the c-myc locus was measured in cells incubated with equitoxic concentrations of araC or FaraA. In araC-treated cells, nascent DNA from the lagging strand was enriched about 5-fold compared with the leading strand. In contrast, FaraA did not induce any replication imbalance. AraC- and FaraA induced changes in the frequency of N-(phosphonacetyl)-l-aspartate (PALA) resistance and the extent of CAD gene amplification were monitored as markers of drug-induced genomic instability. At concentrations that reduced cloning efficiency by 50% (IC(50)), araC increased the frequency of PALA resistance about 4-fold, while FaraA did not have a significant effect on the frequency of PALA resistance. Pretreatment with araC also increased the extent of CAD gene amplification. We propose that the imbalanced DNA synthesis induced by araC leads to the accumulation of Okazaki fragments on the lagging arms and single-stranded DNA regions on the leading arms of replication forks. The formation of these abnormal replication structures was associated with the generation of genomic instability.


Asunto(s)
Antineoplásicos/farmacología , Citarabina/farmacología , ADN de Neoplasias/efectos de los fármacos , Vidarabina/análogos & derivados , Vidarabina/farmacología , Replicación del ADN/efectos de los fármacos , ADN de Neoplasias/biosíntesis , Humanos , Leucemia , Células Tumorales Cultivadas
12.
Cancer Res ; 60(14): 3790-9, 2000 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10919652

RESUMEN

The c-myc gene is frequently deregulated and overexpressed in human cancers, and strategies designed to inhibit c-myc expression in cancer cells may have considerable therapeutic value. The purpose of the present work was to characterize the antigene and antiproliferative activity of a triple helix-forming oligonucleotide (TFO) targeted to a homopurine-homopyrimidine sequence in the P2 promoter of the c-myc gene. The TFO was synthesized with phosphorothioate (PS) internucleotide linkages to confer resistance to intra- and extracellular nucleases. This property is required of oligonucleotides designed for in vivo testing and therapeutic applications. The PS-TFO was found to form triplex DNA with affinity and specificity comparable with that of the corresponding phosphodiester TFO, as shown by gel mobility shift and footprinting assays. Fluorescence microscopy and polyacrylamide gel analysis showed that the fluorescein-labeled PS-TFO accumulated in nuclei of CEM leukemia cells and remained intact for at least 72 h. Incubation of CEM cells with PS-TFO reduced c-myc RNA and protein levels. A single exposure of leukemia cells to the PS-TFO was sufficient to induce dose-dependent growth inhibitory effects. Growth inhibition correlated with accumulation of cells in S phase and with induction of cell death by apoptosis. The PS-TFO was also effective in other leukemia and lymphoma cell lines. Control oligonucleotides had minimal effects in all assays. These data indicate that the c-myc-targeted PS-TFO is an effective antigene and antiproliferative agent, with potential for testing in vivo as a novel approach to cancer therapy.


Asunto(s)
ADN , Leucemia/tratamiento farmacológico , Leucemia/genética , Oligonucleótidos/farmacología , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/inmunología , Antígenos/metabolismo , Apoptosis/efectos de los fármacos , Western Blotting , División Celular/efectos de los fármacos , División Celular/genética , Núcleo Celular/metabolismo , Medios de Contraste/farmacología , ADN Complementario/metabolismo , Dimetilsulfóxido/farmacología , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Exones , Fluoresceína/farmacología , Células HL-60 , Humanos , Linfoma/tratamiento farmacológico , Linfoma/genética , Microscopía Fluorescente , Oligonucleótidos/farmacocinética , ARN/metabolismo , Fase S/efectos de los fármacos , Solventes/farmacología , Factores de Tiempo , Células Tumorales Cultivadas
13.
Biochemistry ; 39(17): 5126-38, 2000 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-10819980

RESUMEN

Triple helix-forming oligonucleotides (TFOs) bind with high affinity and specificity to homopurine-homopyrimidine sequences in DNA and have been shown to inhibit transcription of target genes in various experimental systems. In the present study, we evaluated the ability of 3'-amino-modified phosphodiester TFOs directed to four sites in the c-myc gene to inhibit gene expression and proliferation of human leukemia (CEM, KG-1, and HL-60) and lymphoma (Raji and ST486) cells. GT-rich TFOs were designed to target sequences located either upstream (myc1 and -2) or downstream (myc3 and -4) of the P2 promoter, which is the major c-myc promoter. Myc2, which was directed to a site immediately upstream of this promoter, inhibited c-myc expression and proliferation of CEM cells. The effects of this TFO were sequence- and target-specific, since control oligonucleotides and TFOs directed to other sites were less or not active. Myc2 was also effective in KG-1, HL-60, and Raji cells. In contrast, ST486 cells were more sensitive to myc3, which targets a sequence in intron 1 upstream of the P3 promoter, than myc2. As result of a chromosomal translocation, P3 is the active promoter in ST486 cells. This study demonstrates the activity and specificity of TFOs designed to act as repressors of c-myc gene expression in human leukemia and lymphoma cells. Our results suggest that this is a valid approach to selectively inhibit gene expression and cancer cell growth, and encourage further investigation of its potential applications in cancer therapy.


Asunto(s)
Regulación de la Expresión Génica , Genes myc , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/genética , División Celular/genética , Línea Celular , Humanos , Conformación de Ácido Nucleico , Oligonucleótidos Antisentido/administración & dosificación
14.
Br J Cancer ; 80(7): 971-80, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10362104

RESUMEN

Thiocoraline, a new anticancer agent derived from the marine actinomycete Micromonospora marina, was found to induce profound perturbations of the cell cycle. On both LoVo and SW620 human colon cancer cell lines, thiocoraline caused an arrest in G1 phase of the cell cycle and a decrease in the rate of S phase progression towards G2/M phases, as assessed by using bromodeoxyuridine/DNA biparametric flow cytometric analysis. Thiocoraline does not inhibit DNA-topoisomerase II enzymes in vitro, nor does it induce DNA breakage in cells exposed to effective drug concentrations. The cell cycle effects observed after exposure to thiocoraline appear related to the inhibition of DNA replication. By using a primer extension assay it was found that thiocoraline inhibited DNA elongation by DNA polymerase alpha at concentrations that inhibited cell cycle progression and clonogenicity. These studies indicate that the new anticancer drug thiocoraline probably acts by inhibiting DNA polymerase alpha activity.


Asunto(s)
Antibacterianos/farmacología , Antibióticos Antineoplásicos/farmacología , Depsipéptidos , Péptidos , Bromodesoxiuridina/metabolismo , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , ADN/efectos de los fármacos , ADN/metabolismo , ADN Polimerasa Dirigida por ADN/efectos de los fármacos , Citometría de Flujo , Humanos , Concentración 50 Inhibidora , Factores de Tiempo , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
15.
Biochemistry ; 36(19): 5739-48, 1997 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-9153414

RESUMEN

Recent studies have shown that the anticancer drugs VM-26 and mitoxantrone stabilize preferentially the binding of topoisomerase IIalpha to replicating compared to nonreplicating DNA. To further understand the mechanisms by which cleavable complex-forming topoisomerase II inhibitors interfere with DNA replication, we examined the effects of VM-26 on this process in human leukemia CEM cells. Both the inhibition of DNA synthesis and cell survival were directly related to the total amount of drug-stabilized cleavable complexes formed in VM-26-treated cells. DNA chain elongation was also inhibited in a concentration-dependent fashion in these cells, which suggested that VM-26-stabilized cleavable complexes interfered with the movement of DNA replication forks. To test this hypothesis directly, we monitored replication fork progression at a specific site of VM-26-induced DNA cleavage. A topoisomerase II-mediated cleavage site was detected in the first exon of the c-myc gene in VM-26-treated cells. This cleavage site was downstream of a putative replication origin located in the 5' flanking region of the gene. Replication forks, which moved through this region of the c-myc gene in the 5' to 3' direction, were specifically arrested at this site in VM-26-treated cells, but not in untreated or aphidicolin-treated cells. These studies provide the first direct evidence that a VM-26-stabilized topoisomerase II-DNA cleavable complex acts as a replication fork barrier at a specific genomic site in mammalian cells. Furthermore, the data support the hypothesis that the replication fork arrest induced by cleavable complex-forming topoisomerase II inhibitors leads to the generation of irreversible DNA damage and cytotoxicity in proliferating cells.


Asunto(s)
Daño del ADN , Replicación del ADN/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/farmacología , Leucemia de Células T/tratamiento farmacológico , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Tenipósido/farmacología , ADN-Topoisomerasas de Tipo II/metabolismo , Estabilidad de Enzimas/efectos de los fármacos , Genes myc/efectos de los fármacos , Humanos , Leucemia de Células T/genética , Sustancias Macromoleculares , Inhibidores de Topoisomerasa II , Células Tumorales Cultivadas
16.
Biochemistry ; 35(50): 16354-60, 1996 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-8973211

RESUMEN

Several clinically active anticancer drugs are known to interfere with DNA topoisomerase II activity. However, the importance of the individual alpha (170 kDa) and beta (180 kDa) isozymes as targets of topoisomerase II-active drugs is not clear. To address this question, human CCRF-CEM leukemia cells were incubated with bromodeoxyuridine, and either the nascent DNA or bulk DNA not undergoing replication was purified by immunoprecipitation with an anti-bromodeoxyuridine antibody. The topoisomerase II isozymes that coprecipitated with either the nascent DNA or bulk DNA were analyzed by Western blotting. The alpha isozyme formed complexes with nascent DNA in cells pretreated with either VM-26 or mitoxantrone, while the beta isozyme was only bound to bulk DNA. At moderately cytotoxic concentrations, VM-26 enhanced the binding of topoisomerase II alpha to nascent DNA at least 5.2-fold compared to bulk DNA. However, in VM-26 resistant CEM/VM-1 cells incubated with equitoxic concentrations of VM-26, topoisomerase II alpha complex formation with nascent DNA was decreased at least 5.5-fold compared to bulk DNA. Drug-induced binding of topoisomerase II beta with bulk DNA in CEM/VM-1 cells did not correlate with cytotoxicity. Collectively, these results indicate that the formation of VM-26 stabilized complexes of topoisomerase II alpha with nascent DNA are critical to the development of cytotoxicity, and that resistance of CEM/VM-1 cells to VM-26 is related to impaired formation of these complexes. The results also provide indirect evidence that topoisomerase II alpha is involved in DNA, replication.


Asunto(s)
Antineoplásicos Fitogénicos/toxicidad , Replicación del ADN/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/metabolismo , ADN de Neoplasias/metabolismo , Etopósido/farmacología , Isoenzimas/metabolismo , Antineoplásicos Fitogénicos/metabolismo , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/aislamiento & purificación , ADN de Neoplasias/biosíntesis , ADN de Neoplasias/aislamiento & purificación , Etopósido/metabolismo , Humanos , Isoenzimas/aislamiento & purificación , Cinética , Leucemia-Linfoma Linfoblástico de Células Precursoras , Unión Proteica , Células Tumorales Cultivadas
18.
Cancer Res ; 55(19): 4352-60, 1995 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-7671247

RESUMEN

WEHI-3B/NOVO is a cloned murine leukemia cell line selected for resistance to novobiocin that is cross-resistant to the cytotoxic action of etoposide (VP-16) and to a lesser extent to a variety of other topoisomerase II (topo II)-reactive drugs. We have reported previously (Cancer Res. 52: 2782-2790, 1992) that WEHI-3B/NOVO cells exhibit a pronounced decrease in VP-16 induced DNA-topo II cross-link formation compared to the parental WEHI-3B/S cell line in intact cells, in the absence of a significant difference in the P4 unknotting activity of topo II assayed in nuclear extracts. Because the pattern of cross-resistance was suggestive of a topo II-mediated mechanism, we have ascertained whether a change in topo II can account for the multidrug-resistant phenotype of WEHI-3B/NOVO cells. No differences existed between WEHI-3B/S and WEHI-3B/NOVO cells in topo II mRNA and protein levels, as well as in the amount of topo II associated with the nuclear matrix. Neither sensitive nor resistant cells expressed detectable levels of the MDR1 gene; however, VP-16 accumulation in WEHI-3B/NOVO cells was 3-4-fold less than that present in WEHI-3B/S cells, whereas doxorubicin accumulation was the same in both cell lines. Over the first 60 s, no difference existed in the rate of uptake of VP-16 between parental and resistant cells; however, beyond the first 60 s of incubation, [3H]VP-16 accumulated to a greater extent in parental sensitive cells. Thus, an increased rate of efflux of VP-16 was responsible for the lower steady-state concentration of the drug in resistant cells. The efflux Km for VP-16 in WEHI-3B/NOVO cells was 254.7 microM and the Vmax was 10.4 pmol/s/10(7) cells. In the presence of the inhibitors of energy metabolism, sodium azide and deoxyglucose, the efflux of VP-16 was markedly inhibited; readdition of glucose restored the original efflux rate. Northern blot analyses using the human 10.1 probe for the 3'-terminal region of the multidrug-resistance protein (MRP) cDNA revealed a mRNA species of approximately 6 kb in WEHI-3B/NOVO cells but not in WEHI-3B/S cells. Overexpression was associated with amplification of the cognate gene. To ascertain whether the overexpressed gene in WEHI-3B/NOVO cells was the murine MRP or a different member of the same superfamily of ATP-binding ABC cassette transporters, a 341-bp MRP cDNA probe was generated from a murine genomic library.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Adenosina Trifosfato/farmacología , Antineoplásicos Fitogénicos/farmacocinética , Resistencia a Múltiples Medicamentos/genética , Etopósido/farmacocinética , Leucemia Experimental/metabolismo , Secuencia de Bases , Transporte Biológico , ADN-Topoisomerasas de Tipo II/análisis , Immunoblotting , Datos de Secuencia Molecular , Novobiocina/farmacología , Matriz Nuclear/enzimología , Células Tumorales Cultivadas
19.
Mol Pharmacol ; 47(5): 948-55, 1995 May.
Artículo en Inglés | MEDLINE | ID: mdl-7746281

RESUMEN

Inhibitors of IMP dehydrogenase (EC 1.2.1.14), including mizoribine (Bredinin) and mycophenolic acid, have significant antitumor and immunosuppressive activities. Studies were aimed at determining the mechanism by which intracellular GTP depletion induced by these agents results in inhibition of DNA synthesis. Incubation of human CEM leukemia cells for 2 hr with IC50 concentrations of either mizoribine (4 microM) or mycophenolic acid (0.5 microM) reduced cellular GTP levels an average of 68% or 58%, respectively, compared with the levels in control cells. Under similar conditions, mizoribine and mycophenolic acid decreased the amount of [3H]adenosine incorporated into primer RNA by 75% and 70%, respectively, relative to the untreated controls, but had no significant effect on total RNA synthesis. Repletion of the guanine nucleotide pools by coincubation of CEM cells with guanosine plus 8-aminoguanosine prevented both the inhibition of primer RNA synthesis and the inhibition of tumor cell growth induced by these agents. Additional studies demonstrated that GTP depletion alone was capable of directly inducing inhibition of primer RNA synthesis. Primer RNA synthesis was inhibited an average of 84% in whole-cell lysates that lacked GTP but contained all remaining ribo- and deoxyribonucleoside triphosphates. On an M13 DNA template, RNA-primed DNA synthesis catalyzed by the purified complex of DNA primase (EC 2.7.7.6) and DNA polymerase alpha (EC 2.7.7.7) was decreased an average of 70% in the absence of GTP, compared with synthesis in the presence of 0.5 mM GTP. These results provide evidence that mizoribine and mycophenolic acid inhibit DNA replication by inducing GTP depletion, which suppresses the synthesis of RNA-primed DNA intermediates.


Asunto(s)
ADN de Neoplasias/biosíntesis , Guanosina Trifosfato/metabolismo , IMP Deshidrogenasa/antagonistas & inhibidores , ARN Neoplásico/metabolismo , Adenosina Trifosfato/metabolismo , Supervivencia Celular/efectos de los fármacos , ADN Primasa , ADN de Neoplasias/metabolismo , Humanos , Leucemia/metabolismo , Ácido Micofenólico/farmacología , Nucleosomas/efectos de los fármacos , Nucleosomas/metabolismo , ARN Nucleotidiltransferasas/metabolismo , Ribonucleósidos/farmacología , Células Tumorales Cultivadas
20.
Adv Enzyme Regul ; 35: 265-81, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-7572348

RESUMEN

DNA topoisomerase II is a major protein of the nuclear matrix. The enzyme appears to have a central role in both DNA organization and replication. The importance of nuclear matrix topoisomerase II alpha as a target for certain anticancer agents was evaluated in CEM human leukemia cells. Studies were done to determine the extent to which the alpha (170 kDa) and beta (180 kDa) isozymes of topoisomerase II form covalent enzyme-DNA complexes in whole cells and in the nuclear matrix and nonmatrix fractions of CEM cells that are either sensitive or resistant to topoisomerase II-active anticancer agents. Topoisomerase II alpha was detected in both the high salt-soluble (nonmatrix) and matrix fractions of nuclei from parental CEM cells. Most of the matrix topoisomerase II alpha was tightly bound to DNA in cells incubated with VM-26. In contrast, topoisomerase II beta was detected only in the high salt-soluble (nonmatrix) fraction of the nucleus. The subnuclear distribution of the alpha and beta topoisomerase II isozymes in CEM/VM-1 cells resistant to topoisomerase-active drugs was similar to that in drug-sensitive CEM cells. However, the amount and activity of topoisomerase II alpha in nuclear matrices of CEM/VM-1 cells were decreased 3- to 6-fold relative to that of CEM cells. The differences observed in the subnuclear distribution and DNA binding pattern of the topoisomerase II isozymes support the hypotheses that each isozyme has a distinct cellular function. Furthermore, these results provide evidence that topoisomerase II alpha is the nuclear matrix target for VM-26, and that depletion of the nuclear matrix isozyme contributes to cellular resistance to this anticancer agent.


Asunto(s)
Antineoplásicos/farmacología , ADN-Topoisomerasas de Tipo II/metabolismo , ADN de Neoplasias/metabolismo , Leucemia/enzimología , Matriz Nuclear/enzimología , Tenipósido/farmacología , Adenosina Trifosfato/farmacología , Núcleo Celular/enzimología , Resistencia a Antineoplásicos , Humanos , Isoenzimas/metabolismo , Cloruro de Sodio/farmacología , Inhibidores de Topoisomerasa II , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...