Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Chemother Pharmacol ; 68(1): 193-205, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20886212

RESUMEN

PURPOSE: GTI-2040, a 20-mer phosphorothioate oligonucleotide, was designed to hybridize to the mRNA sequence of human ribonucleotide reductase R2. GTI-2040 has been shown to inhibit human cancer cell proliferation by downregulation of R2 expression in vitro and to significantly inhibit tumor growth in xenograft models of human cancer in mice. As part of the safety evaluation for human clinical trials, the toxicity and toxicokinetics of GTI-2040 were determined in Sprague-Dawley rats and rhesus monkeys. METHODS: GTI-2040 was administered to rats at 2, 10, and 50 mg/kg/day by bolus intravenous injection every second day for 21 days with a 21-day recovery. In monkeys, an acute study was performed with single, escalating doses of GTI-2040 ranging from 10 to 80 mg/kg given as a 24-h continuous intravenous infusion. As well, a 21-day, continuous intravenous infusion study with GTI-2040 was conducted in monkeys at 2, 10, and 50 mg/kg/day, with a 3-week recovery. Blood sampling was done to measure GTI-2040 plasma concentrations, metabolites, and pharmacokinetic parameters, and tissues were collected to assess the distribution of GTI-2040 and/or metabolites. RESULTS: The toxicities of GTI-2040 in both rats and monkeys were typical for the phosphorothioate oligonucleotide class of compounds. In monkeys, there was a dose-related increase in GTI-2040 plasma levels with concomitant increase in complement activation and prolongation of activated partial thromboplastin time. In both rats and monkeys, the tissues having the highest concentrations of GTI-2040 (kidney, liver, spleen) had the largest dose-related toxic effects. Adverse effects were diminished or absent in the recovery animals. CONCLUSIONS: GTI-2040 was well tolerated when infused over 24 h at doses up to 80 mg/kg in monkeys. In rats and monkeys, GTI-2040 was reasonably well tolerated and showed reversible toxicities when administered at doses up to 50 mg/kg/day for 21 days. The no observed adverse effect dose level for GTI-2040 in both animal species was 2 mg/kg/day. There were no apparent sequence-specific effects related to the interaction of GTI-2040 with the R2 component of the mRNA expressing ribonucleotide reductase.


Asunto(s)
Antineoplásicos/farmacocinética , Antineoplásicos/toxicidad , Oligodesoxirribonucleótidos/farmacocinética , Oligodesoxirribonucleótidos/toxicidad , Oligonucleótidos Fosforotioatos/farmacocinética , Oligonucleótidos Fosforotioatos/toxicidad , Ribonucleótido Reductasas/genética , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/sangre , Secuencia de Bases , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Macaca mulatta , Masculino , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/sangre , Oligonucleótidos Fosforotioatos/administración & dosificación , Oligonucleótidos Fosforotioatos/sangre , ARN Mensajero/química , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Ribonucleótido Reductasas/metabolismo
2.
Bioorg Med Chem Lett ; 20(12): 3518-20, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20483613

RESUMEN

A new series of antimicrobial derivatives [3-(4,5-diaryl-1H-imidazol-2-yl)-1H-indole)] have been synthesized with potent activity against strains of Staphylococcus aureus, including methicillin-resistant strains (MRSA). Compound 17 [3-(4,5-bis(4-fluorophenyl)-1H-imidazol-2-yl)-5-bromo-1H-indole], the most active derivative was shown to inhibit the growth of all Gram-positive strains tested, including vancomycin resistant Enterococcus faecalis and Enterococcus faecium with no activity against Gram-negative bacteria.


Asunto(s)
Antiinfecciosos/síntesis química , Indoles/síntesis química , Indoles/farmacocinética , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Antiinfecciosos/farmacología , Enterococcus faecalis/efectos de los fármacos , Enterococcus faecium/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Vancomicina/farmacología
3.
Cancer Immunol Immunother ; 59(6): 805-17, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20012860

RESUMEN

Interleukin-17E (IL-17E) belongs to a novel family of cytokines that possess significant homology to IL-17. IL-17E has potent inflammatory effects in vitro and in vivo. Overexpression of IL-17E in mice results in a T helper-2 (Th2)-type immune response, which includes the expansion of eosinophils through the production of IL-5, and elevated gene expression of IL-4 and IL-13 in multiple tissues. In this study, we show that IL-17E has antitumor activity in vivo, a previously unrecognized function of IL-17E. Antitumor efficacy of IL-17E was examined in a variety of human tumor xenograft models, including melanoma, breast, lung, colon, and pancreatic cancers. Injection of recombinant IL-17E every other day resulted in significant antitumor activity in these tumor models. In addition, the combination of IL-17E with chemotherapy or immunotherapy agents showed an enhanced antitumor efficacy in human tumor xenograft models in mice as compared to either agent alone. Antitumor activity was demonstrated using different routes of administration, including intraperitoneal, intravenous, and subcutaneous injection. Anticancer activity was shown for both mouse and human forms of IL-17E, which have a high degree of sequence identity. Tumor-bearing mice treated with IL-17E showed a significant increase in serum levels of IL-5 and increased numbers of eosinophils in peripheral blood compared to the control group. Spleens isolated from IL-17E-treated mice showed a significant increase in eosinophils that correlated with antitumor activity of IL-17E in a dose-response manner. Finally, we demonstrate that B cells are necessary for IL-17E-mediated antitumor activity and that IL-17E was found to activate signaling pathways in B cells in vitro. Taken together, these data demonstrate that IL-17E has antitumor activity in vivo, and support further investigation of the potential clinical use of IL-17E as an anticancer agent.


Asunto(s)
Linfocitos B/metabolismo , Interleucina-17/metabolismo , Interleucina-5/metabolismo , Neoplasias Experimentales/metabolismo , Animales , Antineoplásicos/farmacología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/patología , Eosinofilia/inmunología , Femenino , Células HT29 , Humanos , Procesamiento de Imagen Asistido por Computador , Interleucina-17/genética , Interleucina-17/farmacología , Interleucina-5/genética , Interleucina-5/inmunología , Ratones , Ratones Desnudos , Ratones SCID , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Transducción de Señal , Células Th2/efectos de los fármacos , Células Th2/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Mol Cancer Ther ; 8(9): 2586-96, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19755513

RESUMEN

ML-133 is a novel small molecule with potent antiproliferative activity, as shown in cancer cell lines and in a human colon tumor xenograft model. ML-133 reduces the concentration of intracellular labile zinc in HT-29 colon cancer cells, leading to induction of the Krüppel-like factor 4 transcription factor. Krüppel-like factor 4 displaces the positive regulator SP1 from the cyclin D1 promoter, thereby negatively regulating the expression of cyclin D1 and promoting the G(1)-S phase arrest of cell proliferation. The antiproliferative and antitumor activity of ML-133 described in the present study suggests modulation of intracellular zinc homeostasis as a potential strategy for the treatment of several cancer types, and ML-133 represents a promising new class of antitumor agents that deserves further development.


Asunto(s)
Antineoplásicos/farmacología , División Celular/efectos de los fármacos , Homeostasis/efectos de los fármacos , Imidazoles/farmacología , Fenantrolinas/farmacología , Zinc/metabolismo , Secuencia de Bases , Western Blotting , Ciclo Celular , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Ciclina D1/genética , Cartilla de ADN , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Células HT29 , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/biosíntesis , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Factor de Transcripción Sp1/metabolismo , Regulación hacia Arriba/efectos de los fármacos
5.
Int J Oncol ; 34(1): 33-42, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19082475

RESUMEN

GTI-2040, an antisense oligonucleotide targeting the small subunit of ribonucleotide reductase, acts as an anti-tumor agent in animal models of human cancer. In the present study, the anti-tumor activity of GTI-2040, in combination with interferon alpha (IFNalpha) was investigated against human renal cell carcinoma tumors xenografted into mice. The human renal cell carcinoma cell lines, Caki-1 and A498 were sensitive to IFNalpha both in vitro and when implanted into mice. In combination with GTI-2040 there were cooperative effects at intermediate doses of the two agents and complete tumor regression at higher combination doses. A control oligonucleotide was not effective as a monotherapy and did not improve the efficacy of IFNalpha. The effect of combination treatment on apoptosis and proliferation of tumor cells, isolated from xenografted tumors, was examined by histochemistry. GTI-2040 increased the percentage of cells undergoing apoptosis with a concomitant decrease in proliferation. IFNalpha alone had no effect but in combination with GTI-2040 resulted in increased apoptosis and decreased proliferation compared to GTI-2040 alone. Taken together these results expand the potential clinical applications of GTI-2040 to include combination therapy with IFNalpha.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Células Renales/tratamiento farmacológico , Interferón-alfa/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Oligodesoxirribonucleótidos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Proliferación Celular/efectos de los fármacos , Quimioterapia Combinada , Terapia Genética , Humanos , Técnicas para Inmunoenzimas , Neoplasias Renales/genética , Neoplasias Renales/patología , Ratones , Ratones SCID , Ribonucleótido Reductasas/antagonistas & inhibidores , Ribonucleótido Reductasas/genética , Trasplante Heterólogo , Células Tumorales Cultivadas
6.
Bioorg Med Chem Lett ; 19(1): 104-7, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19027297

RESUMEN

A series of 11-phenyl-[b,e]-dibenzazepine compounds were synthesized and shown to be inhibitors of tumor cell proliferation with IC(50) values ranging from submicromolar to micromolar concentrations. Flow cytometric analyses of several active compounds demonstrated inhibition of cell cycle progression at the G(0)-G(1) phase transition resulting in G(0)-G(1) arrest.


Asunto(s)
Antineoplásicos/síntesis química , Dibenzazepinas/síntesis química , Dibenzazepinas/farmacología , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Fase G1 , Humanos , Concentración 50 Inhibidora , Fase de Descanso del Ciclo Celular , Relación Estructura-Actividad
7.
Cancer Immunol Immunother ; 57(12): 1757-69, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18351336

RESUMEN

Virulizin has demonstrated strong antitumor efficacy in a variety of human tumor xenograft models including melanoma, pancreatic cancer, breast cancer, ovarian cancer and prostate cancer. Our previous studies have demonstrated that macrophages, NK cells, and cytokines are important in the antitumor mechanism of Virulizin. Virulizin treatment of tumor bearing mice results in the expansion as well as increased activity of monocytes/macrophages and production of cytokines IL-12 and TNFalpha and activation of NK cells. In this study we show that the inflammatory cytokine IL-17E (IL-25) is induced by Virulizin treatment and is part of its antitumor mechanism. IL-17E is a proinflammatory cytokine, which induces a T(H)2 type immune response, associated with eosinophil expansion and infiltration into mucosal tissues. IL-17E was increased in sera of Virulizin-treated mice bearing human melanoma xenografts, compared to saline-treated controls, as shown by 2D gel electrophoresis and ELISA. Treatment of splenocytes in vitro with Virulizin resulted in increased IL-17E mRNA expression, which peaked between 24 and 32 h post-stimulation. Both in vitro and in vivo experiments demonstrated that B cells produced IL-17E in response to Virulizin treatment. Furthermore, Virulizin treatment in vivo resulted in increased blood eosinophilia and eosinophil infiltration into tumors. Finally, injection of recombinant IL-17E showed antitumor activity towards xenografted tumors, which correlated with increased eosinophilia in blood and tumors. Taken together, these results support another antitumor mechanism mediated by Virulizin, through induction of IL-17E by B cells, leading to recruitment of eosinophils into tumors, which may function in parallel with macrophages and NK cells in mediating tumor destruction.


Asunto(s)
Antineoplásicos/uso terapéutico , Quimiotaxis de Leucocito/efectos de los fármacos , Eosinófilos/efectos de los fármacos , Interleucina-17/biosíntesis , Melanoma Experimental/tratamiento farmacológico , Extractos de Tejidos/uso terapéutico , Adyuvantes Inmunológicos/uso terapéutico , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Bilis , Western Blotting , Electroforesis en Gel Bidimensional , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Int Immunopharmacol ; 7(10): 1350-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17673150

RESUMEN

Virulizin, a novel biological response modifier, has demonstrated broad antitumor efficacy in a variety of human tumor xenograft models including melanoma, pancreatic cancer, breast cancer, ovarian cancer and prostate cancer. Previous studies have demonstrated a significant role of macrophages and NK cells in the antitumor mechanism of Virulizin. Increased activity and expansion of macrophages and NK cells has been observed in mice treated with Virulizin. In the present study, the effects of Virulizin on TNFalpha expression were investigated in vitro and in vivo. CD-1 nude mice were treated with Virulizin daily for 5 days. Quantitative RT-PCR demonstrated that the level of TNFalpha mRNA increased in peritoneal macrophages isolated from Virulizin-treated mice as compared to the control group. An increase in TNFalpha protein expression was also observed, as assessed by flow cytometric analysis. Increased levels of TNFalpha mRNA were seen in human tumor xenografts following treatment of tumor-bearing mice with Virulizin. In the presence of LPS, Virulizin also stimulated TNFalpha protein secretion and mRNA expression in human monocytic U937 cells and mouse macrophage RAW264.7 cells in vitro in a time- and dose-dependent manner. U937 cells treated with Virulizin showed a significantly enhanced cytotoxicity that was eliminated upon neutralization of TNFalpha. Virulizin also induced the phosphorylation of IkappaB, suggesting that induction of TNFalpha expression by Virulizin is mediated by activation of NFkappaB. The results indicate that Virulizin-induced TNFalpha expression contributes to modulation of immune responses and antitumor activities.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Monocitos/efectos de los fármacos , Extractos de Tejidos/farmacología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Bilis , Línea Celular , Línea Celular Tumoral , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunoterapia , Macrófagos Peritoneales/inmunología , Melanoma/inmunología , Ratones , Ratones Desnudos , Monocitos/inmunología , Trasplante de Neoplasias , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/genética , Células U937
9.
Anticancer Drugs ; 18(4): 377-88, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17351390

RESUMEN

RNA interference, a posttranscriptional gene-silencing mechanism, has received considerable attention for its potential as a new therapeutic strategy to treat human diseases and conditions including cancer. Various studies have supported a role for the R2 subunit of ribonucleotide reductase in cancer progression and metastasis. Short interfering siRNA 1284 was designed to target R2. In vitro studies, in which three different human tumor cell lines (A498, HT-29 and A2058) were transfected with short interfering siRNA 1284, demonstrate sequence-specific down-regulation of R2, which coincides with a decrease in cell proliferation, and cell cycle inhibition. In vivo studies with xenograft mouse models, generated from the same tumor cell lines, indicate that treatment with short interfering siRNA 1284 leads to inhibition of tumor growth and this effect was found to be dose dependent. Taken together, these results suggest that short interfering siRNA 1284, targeting R2, has great potential to serve as a therapeutic agent towards the treatment of human cancers.


Asunto(s)
Neoplasias/terapia , ARN Interferente Pequeño/uso terapéutico , Ribonucleótido Reductasas/biosíntesis , Animales , Northern Blotting , Western Blotting , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Densitometría , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Células HT29 , Humanos , Ratones , Ratones SCID , Trasplante de Neoplasias , Neoplasias/genética , Ribonucleótido Reductasas/genética , Fase S/efectos de los fármacos , Transfección , Trasplante Heterólogo
10.
Anticancer Drugs ; 17(2): 143-54, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16428932

RESUMEN

Human thioredoxin has been implicated in cancer as a growth stimulator through regulation of DNA replication and growth factor activity, as a modulator of transcription factor activity, and as an inhibitor of apoptosis. In the present study, the steady-state level of thioredoxin protein was examined in a number of cancer cell lines. Interestingly, thioredoxin expression is elevated in a variety of human tumor cell lines compared with normal cell lines. The altered expression of thioredoxin in tumor cells suggests it may be a target in the development of novel therapeutic agents for the treatment and prevention of cancer. Further to this possibility, 26 phosphorothioate antisense oligodeoxynucleotides (PS-AS-ODNs) were evaluated for the ability to inhibit thioredoxin expression in cell culture. One PS-AS-ODN, GTI-2601, specifically reduced the levels of thioredoxin mRNA and protein, exhibited potent anti-proliferative effects on colony formation in vitro, and had anti-tumor effects in human tumor xenograft mouse models in vivo. Sequence-specific decreases in thioredoxin expression levels were accompanied by significant suppression of tumor growth in mice. Taken together, these data suggest that thioredoxin may be a useful target for developing PS-AS-ODNs as drug candidates against human cancer.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Oligonucleótidos Antisentido/uso terapéutico , Tiorredoxinas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Femenino , Humanos , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Neoplasias/patología , Tiorredoxinas/metabolismo
11.
Int J Oncol ; 28(2): 469-78, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16391803

RESUMEN

GTI-2501 is a 20-mer oligonucleotide that is complementary to a coding region in the mRNA of R1, the large subunit of ribonucleotide reductase (RNR). In vitro studies, have demonstrated that GTI-2501 decreases mRNA and protein levels of R1 in a sequence-specific and dose-dependent manner. Furthermore, GTI-2501 inhibits the growth of human lung, liver, ovary, brain, melanoma, breast and pancreatic tumor cells in colony forming assays. In vivo studies have shown that GTI-2501 significantly inhibits growth of human colon, pancreas, lung, breast, renal, ovarian, melanoma, brain glioblastoma-astrocytoma, and prostatic tumors in CD-1 nude, Balb/c nude and/or SCID mice. GTI-2501 treatment caused total regression of human breast and renal tumor xenografts in mice. These effects are not observed with a scrambled control oligonucleotide containing the same base content but not complementary to R1. GTI-2501 specifically inhibits metastasis of human melanoma cells to the lungs in CD-1 athymic nude mice and prolongs the survival of mice bearing human lymphoma. Taken together these results suggest that an antisense mechanism of action is responsible for growth inhibition in vitro and in vivo and that GTI-2501 can act as a selective and specific anti-tumor agent.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Experimentales/prevención & control , Oligonucleótidos Antisentido/farmacología , Ribonucleótido Reductasas/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células HT29 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones SCID , Neoplasias Experimentales/patología , Oligonucleótidos , ARN Mensajero/metabolismo , Ribonucleótido Reductasas/genética , Ribonucleótido Reductasas/metabolismo , Ensayo de Tumor de Célula Madre
12.
Cancer Chemother Pharmacol ; 58(3): 306-18, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16333677

RESUMEN

PURPOSE: A cholesterol-free liposome formulation formed from mixtures of egg phosphatidylcholine (ePC) and poly (ethylene glycol) conjugated distearoylphosphatidylethanolamine (DSPE-PEG 2000) was optimized and evaluated for delivery of a novel anti-cancer agent ML220 (2-(5-bromo-1H-indol-3-yl)-1H-phenanthro [9,10-d] imidazole). RESULTS AND DISCUSSION: ML220 is highly lipophilic with a water solubility of 0.14 mug/ml and calculated log P of 5.69. The ML220-loaded liposomes had a unimodal size-distribution and a mean diameter of 89 nm. The drug to lipid ratio in the formulation was 1:3.5 (mol:mol) and the drug loading efficiency was 83% providing a more than 50,000-fold increase in the water solubility of ML220. The formulation was demonstrated to be stable in vitro at 37 degrees C for over 2 weeks with a delayed drug release profile. Evaluation of the subacute toxicity of the liposome formulated drug in C3H mice revealed no overt signs of toxicity. Also, a biexponential drug plasma concentration pattern was found upon evaluation of the pharmacokinetics in Balb/C mice. The in vivo evaluation of the anti-cancer activity in a human colon HT29 carcinoma model revealed a significant delay in tumor growth. CONCLUSION: Overall, the ePC/DSPE-PEG liposomes were demonstrated to be a suitable delivery system for ML220. These studies also highlight the potential of cholesterol-free liposomes as a formulation strategy for highly lipophilic drugs.


Asunto(s)
Antineoplásicos , Neoplasias Experimentales/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Composición de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Estabilidad de Medicamentos , Femenino , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Imidazoles/administración & dosificación , Imidazoles/efectos adversos , Imidazoles/farmacocinética , Imidazoles/uso terapéutico , Liposomas , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Desnudos , Tamaño de la Partícula , Fenantrenos/administración & dosificación , Fenantrenos/efectos adversos , Fenantrenos/farmacocinética , Fenantrenos/uso terapéutico , Fosfatidilcolinas/química , Polietilenglicoles/química , Solubilidad , Distribución Tisular
13.
J Clin Lab Anal ; 19(5): 182-8, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16170815

RESUMEN

One major problem associated with collecting whole blood from patients for use as a source of RNA in gene expression studies is that the RNA degrades during collection and storage. Preservation of RNA quality is vital in such studies because the stability of the RNA ultimately affects analysis of gene expression. In this study the PAXgene blood collection system was compared with a standard erythrocyte lysis method for isolating RNA from blood samples. The methods were compared in terms of RNA yield, RNA stabilization, and DNA contamination. The study also included the downstream application to RT-PCR analysis for relative mRNA expression levels of the ribonucleotide reductase subunits R1 and R2. The results show that blood collection in conventional collection tubes, and leukocyte isolation by erythrocyte lysis lead to significant degradation of RNA. Our findings confirm the ability of PAXgene to stabilize RNA in whole blood; however, RNA extracted by the PAXgene method contained significant DNA contamination. Given the low basal expression of the target genes analyzed in this study, contaminating DNA could potentially affect accurate interpretation of RT-PCR data. As a result, the PAXgene protocol was optimized to include off-column DNase treatments, which yielded high-quality RNA suitable for gene expression studies. Furthermore, the results suggest that RNA isolation with PAXgene is advantageous compared to traditional extraction methods for RT-PCR analysis of large or different-sized amplicons.


Asunto(s)
Recolección de Muestras de Sangre/métodos , Perfilación de la Expresión Génica/métodos , ARN/sangre , Actinas/biosíntesis , Desoxirribonucleasas , Estabilidad de Medicamentos , Humanos , Desnaturalización de Ácido Nucleico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleótido Reductasas/biosíntesis
14.
Cancer Immunol Immunother ; 54(11): 1115-26, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15891881

RESUMEN

Virulizin, a novel biological response modifier, has demonstrated significant antitumor efficacy in a variety of human tumor xenograft models including melanoma, pancreatic cancer, breast cancer, ovarian cancer and prostate cancer. The significant role of macrophages and NK (Natural killer) cells was implicated in the antitumor mechanism of Virulizin where expansion as well as increased activity of macrophages and NK cells were observed in mice treated with Virulizin. Depletion of macrophages compromised Virulizin-induced NK1.1+ cell infiltration into xenografted tumors and was accompanied by reduced antitumor efficacy. In the present study, involvement of macrophages in NK cell activation was investigated further. We found that depletion of NK cells in CD-1 nude mice by anti-ASGM1 antibody significantly compromised the antitumor activity of Virulizin. Cytotoxicity of NK cells isolated from Virulizin-treated mice was enhanced against NK-sensitive YAC-1 cells and C8161 human melanoma cells, but not against NK-insensitive P815 cells. An increased level of IL-12beta was observed in the serum of mice treated with Virulizin. IL-12 mRNA and protein levels were also increased in peritoneal macrophages isolated from Virulizin-treated mice. Moreover, Virulizin-induced cytotoxic activity of NK cells isolated from the spleen was abolished when an IL-12 neutralizing antibody was co-administered. In addition, depletion of macrophages in mice significantly impaired Virulizin-induced NK cell cytotoxicty. Taken together, the results suggest that Virulizin induces macrophage IL-12 production, which in turn stimulates NK cell-mediated antitumor activity.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Interleucina-12/genética , Células Asesinas Naturales/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Animales , Bilis , Línea Celular Tumoral , Citotoxicidad Inmunológica , Femenino , Humanos , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Ratones , Trasplante de Neoplasias , ARN Mensajero/análisis , Extractos de Tejidos , Trasplante Heterólogo
15.
Cancer Immunol Immunother ; 54(3): 229-42, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15378281

RESUMEN

Previous studies have demonstrated antitumor efficacy of Virulizin in several human tumor xenograft models and a critical role for macrophages in the antitumor mechanism of Virulizin. Although there is growing support for an immune stimulatory mechanism of action for Virulizin, the details remain to be elucidated. The aim of this study was to determine whether infiltration of natural killer (NK) cells into xenografted tumors is altered by Virulizin treatment, and whether such alterations contribute to the antitumor activity of Virulizin. Immunohistochemical analysis demonstrated that xenografted tumors from Virulizin-treated mice had an increase in infiltration of F4/80(+) (macrophages) and NK1.1(+) (NK) cells. The increase in NK1.1(+) cell infiltration occurred at an early stage of Virulizin treatment, which correlated with an early sign of apoptosis. In addition, Virulizin resulted in an increase in the number of NK cells in the spleens, and NK cells isolated from the spleen exhibited increased cytotoxicity to tumor cells in vitro. In NK cell-deficient SCID-beige mice, the antitumor activity of Virulizin was compromised, providing additional support to the hypothesis that NK cells are necessary for inhibition of tumor growth by Virulizin. Finally, depletion of macrophages resulted in the loss of Virulizin-induced increase in NK1.1(+) cell infiltration into xenografted tumors, suggesting the involvement of macrophages in NK cell infiltration into tumors. Taken together, these results strongly support a mechanism in which Virulizin stimulates a sustained expansion and infiltration of NK cells and macrophages into tumors with subsequent activation of NK cells that is responsible for the observed antitumor activity.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antineoplásicos/farmacología , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Animales , Apoptosis , Bilis , Línea Celular Tumoral , Femenino , Citometría de Flujo , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Células Asesinas Naturales/citología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones SCID , Trasplante de Neoplasias , Péptidos/química , Factores de Tiempo , Extractos de Tejidos
16.
J Pharmacol Exp Ther ; 308(2): 538-46, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14610220

RESUMEN

Although clotrimazole (CLT), an antifungal drug, inhibits tumor cell proliferation and angiogenesis, its clinical application is hampered by significant hepatotoxicity due to the presence of an imidazole moiety. In our attempts to develop CLT analogs that are devoid of imidazole and are as efficacious as CLT, one pharmacophore designated NC381 was generated and shown to inhibit tumor cell growth via a mechanism similar to that of CLT. In vitro, treatment of NCI-H460 nonsmall cell lung cancer (NSCLC) cells with NC381 inhibited growth in a time-dependent manner. Flow cytometric analysis demonstrated that the decrease in cell growth was associated with inhibition of cell cycle progression at the G(1)-S phase transition, resulting in G(0)-G(1) arrest. There was a concomitant inhibition of cyclin D1 expression and subsequent reduction in the formation of the cyclin D1-CDK4 complex. Consistent with a decrease in the cyclin D1-CDK4 complex, NC381 treatment resulted in significant inhibition of pRb phosphorylation. There also were changes in the activity of cell cycle-related proteins, including p16(Ink4) and p27(Kip1). Together, these results are consistent with a model in which NC381 arrests cell cycle progression via inhibition of the pathway that promotes exit from the G(1) phase of the cell cycle. Furthermore, the clinical applicability of NC381 was evaluated in an in vivo murine xenograft model of human NSCLC (NCI-H460). NC381 treatment resulted in significant inhibition of tumor growth. Given the poor prognosis and the limited treatment options available, the present results underscore the potential of NC381 in the treatment of human NSCLC.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Ciclo Celular/metabolismo , Clotrimazol/farmacología , Fase G1/efectos de los fármacos , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Clotrimazol/análogos & derivados , Clotrimazol/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fosforilación/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Bioorg Med Chem Lett ; 14(2): 347-50, 2004 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-14698156

RESUMEN

Clotrimazole (CLT) 1, a synthetic anti-fungal imidazole derivative, inhibits tumor cell proliferation and angiogenesis. In the current study, flow cytometric analysis demonstrated that the decrease in tumor cell growth by CLT 1 was associated with inhibition of cell cycle progression at the G(1)-S phase transition, resulting in G(0)-G(1) arrest. A series of CLT 1 analogues has been generated in order to develop CLT 1 derivatives that are devoid of the imidazole moiety which is responsible for the hepatoxicity associated with CLT 1 while retaining CLT 1 efficacy. The majority of these analogues demonstrate in vitro antiproliferative activity ranging from submicromolar to micromolar concentrations.


Asunto(s)
Inhibidores de Crecimiento/química , Inhibidores de Crecimiento/farmacología , Metano/análogos & derivados , Metano/farmacología , Animales , División Celular/efectos de los fármacos , División Celular/fisiología , Línea Celular Tumoral , Clotrimazol/análogos & derivados , Clotrimazol/farmacología , Humanos , Ratones , Células 3T3 NIH
18.
Int J Oncol ; 23(5): 1341-6, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14532975

RESUMEN

Virulizin, a novel biological response modifier (BRM), has been demonstrated to have a high level of anti-tumor activity against pancreatic cancer and melanoma in many clinical trials and preclinical studies. However, its anti-tumor mechanism has not been fully elucidated. The purpose of this study was to define the mechanism of Virulizin anti-tumor activity in cultures and in a murine xenograft model. The presence of Virulizin stimulated in a dose-dependent manner the cytolytic activity against tumor cells by splenocytes and macrophages, but not by non-adherent splenocytes. The cytotoxic activity of macrophages was significantly increased (approximately 5-fold) in cultures containing 2.5% of Virulizin compared to that of cultures without Virulizin (p<0.001). An increase of 21% in the protease secretion was observed in Virulizin (2.5%)-stimulated macrophages compared to PBS-treated cells (p<0.0001). Moreover, the anti-tumor efficacy of Virulizin observed in CD-1 nude mice was abrogated in mice that were depleted of macrophages, thus stimulation of macrophages may be one mechanism through which Virulizin acts. These results suggest that macrophages may play a critical role in the anti-tumor activity of Virulizin.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antineoplásicos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Animales , Bilis , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Endopeptidasas/metabolismo , Femenino , Citometría de Flujo , Humanos , Melanoma/tratamiento farmacológico , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Pancreáticas/tratamiento farmacológico , Bazo/citología , Factores de Tiempo , Extractos de Tejidos
19.
Clin Cancer Res ; 9(12): 4553-61, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-14555530

RESUMEN

Ribonucleotide reductase is the enzyme responsible for the reduction of ribonucleotides to their corresponding deoxyribonucleotides for DNA synthesis. Ribonucleotide reductase is a multisubunit complex containing two polypeptides, R1 and R2. In addition to catalytic and allosteric regulatory functions, the R1 subunit appears to act as a novel tumor suppressor. Previous studies demonstrated that overexpression of mouse R1 resulted in suppression of tumorigenicity and metastatic potential, whereas expression of antisense RNA, complementary to R1 mRNA, increased anchorage-independent growth of ras-transformed NIH 3T3 cells. The current study investigated the potential of R1 gene therapy for human cancer using a recombinant adenovirus encoding the human R1 gene (rAd5-R1). Recombinant viruses were constructed by FLP-mediated site-specific recombination and demonstrated high infectivity of a human colon carcinoma cell line (Colo320 HRS), as assessed by expression of a viral encoded beta-Gal gene (rAd5-LacZ). R1mRNA and protein were overexpressed in Colo320 HRS cells infected with rAd5-R1 compared with untreated or rAd5-LacZ-infected cells. Infection with rAd5-R1 inhibited Colo320 HRS cell proliferation, in vitro, in a time- and dose-dependent manner. When Colo320 HRS cells were treated with rAd5-R1, before injection into CD-1 mice, there was complete inhibition of tumor growth compared with treatment with rAd5-LacZ. Furthermore, intratumoral injection of rAd5-R1 into Colo320 HRS tumor xenografts inhibited tumor growth in CD-1 mice compared with rAd5-LacZ treated mice (P = 0.0001). These results demonstrate gene-specific antitumor effects of R1 and suggest that rAd5-R1 gene therapy has the potential to improve currently available treatments for colon cancer.


Asunto(s)
Adenocarcinoma/terapia , Adenoviridae/genética , Neoplasias del Colon/terapia , Terapia Genética , Ribonucleótido Reductasas/genética , Adenocarcinoma/enzimología , Adenocarcinoma/patología , Animales , Northern Blotting , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Femenino , Vectores Genéticos , Humanos , Ratones , Ratones Desnudos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribonucleósido Difosfato Reductasa , Transfección , Células Tumorales Cultivadas
20.
Cancer Res ; 63(11): 2802-11, 2003 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12782585

RESUMEN

GTI-2040 is a 20-mer oligonucleotide that is complementary to a coding region in the mRNA of the R2 small subunit component of human ribonucleotide reductase. In vitro studies using a number of human tumor cell lines have demonstrated that GTI-2040 decreases mRNA and protein levels of R2 in a sequence- and target-specific manner. In vivo studies have shown that GTI-2040 significantly inhibits growth of human colon tumors (adenocarcinoma), pancreatic tumors (adenocarcinoma), liver tumors, lung tumors, breast tumors (adenocarcinoma), renal tumors, ovarian tumors (adenocarcinoma), melanoma, brain glioblastoma-astrocytoma, prostatic tumors, and cervical tumors in nude and/or severe combined immunodeficient mice. Antitumor effects were not observed with an oligonucleotide containing four mismatches to the R2 sequence or with a scrambled sequence containing the same base content but not complementary to R2. This suggests that an antisense mechanism is responsible for the in vivo observations. In addition to tumor growth assays, GTI-2040 was tested in a murine model of human lymphoma. Treatment of severe combined immunodeficient mice bearing Burkitt's lymphoma with GTI-2040, but not control oligonucleotides, greatly extended the survival of mice, and survival extended well beyond the treatment period. Finally, GTI-2040 specifically inhibits metastasis of human melanoma cells to the lungs in nude mice. Taken together, the results of these studies indicate that GTI-2040 can act as a selective and specific anticancer agent against a broad range of human tumors.


Asunto(s)
Neoplasias/tratamiento farmacológico , Oligonucleótidos Antisentido/farmacología , Ribonucleótido Reductasas/antagonistas & inhibidores , Animales , Linfoma de Burkitt/tratamiento farmacológico , Linfoma de Burkitt/enzimología , Linfoma de Burkitt/inmunología , Islas de CpG/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Femenino , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/enzimología , Fibrosarcoma/secundario , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/secundario , Melanoma/tratamiento farmacológico , Melanoma/enzimología , Melanoma/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones SCID , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/inmunología , Oligodesoxirribonucleótidos , Oligonucleótidos Antisentido/genética , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ribonucleótido Reductasas/biosíntesis , Ribonucleótido Reductasas/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA